WO2004048343A1 - Chk-, pdk- and akt-inhibitory pyrimidines, their production and use as pharmaceutical agents - Google Patents

Chk-, pdk- and akt-inhibitory pyrimidines, their production and use as pharmaceutical agents Download PDF

Info

Publication number
WO2004048343A1
WO2004048343A1 PCT/EP2003/013443 EP0313443W WO2004048343A1 WO 2004048343 A1 WO2004048343 A1 WO 2004048343A1 EP 0313443 W EP0313443 W EP 0313443W WO 2004048343 A1 WO2004048343 A1 WO 2004048343A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino
phenyl
bromo
pyrimidinyl
alkyl
Prior art date
Application number
PCT/EP2003/013443
Other languages
French (fr)
Inventor
Judi Bryant
Monica Kochanny
Shendong Yuan
Seock-Kyu Khim
Brad Buckman
Damian Arnaiz
Ulf Bömer
Hans Briem
Peter Esperling
Christoph Huwe
Joachim Kuhnke
Martina SCHÄFER
Lars Wortmann
Dirk Kosemund
Emil Eckle
Richard Feldman
Gary Phillips
Original Assignee
Schering Aktiengesellschaft
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to BR0316680-5A priority Critical patent/BR0316680A/en
Priority to MXPA05005547A priority patent/MXPA05005547A/en
Priority to YUP-2005/0363A priority patent/RS20050363A/en
Priority to JP2004554522A priority patent/JP2006508997A/en
Priority to EP03780086A priority patent/EP1565446A1/en
Priority to AU2003288198A priority patent/AU2003288198A1/en
Priority to NZ539823A priority patent/NZ539823A/en
Priority to CA002502970A priority patent/CA2502970A1/en
Application filed by Schering Aktiengesellschaft filed Critical Schering Aktiengesellschaft
Priority to EA200500721A priority patent/EA200500721A1/en
Publication of WO2004048343A1 publication Critical patent/WO2004048343A1/en
Priority to IL168102A priority patent/IL168102A0/en
Priority to IS7881A priority patent/IS7881A/en
Priority to NO20053144A priority patent/NO20053144L/en
Priority to HR20050601A priority patent/HRP20050601A2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/47One nitrogen atom and one oxygen or sulfur atom, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/30Halogen atoms or nitro radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/50Three nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D411/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen and sulfur atoms as the only ring hetero atoms
    • C07D411/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen and sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D411/12Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen and sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Definitions

  • This invention relates to pyrimidine derivatives, their production as well as their use as medications for treating various diseases.
  • Chks checkpoint kinases
  • Akts protein kinases B
  • Pdks phosphoinositide-dependent kinases
  • Akts and Pdks may be involved in common signal transduction pathways.
  • Preferential inhibitors of the Chks and Akts and/or Pdks, particularly of Pdk1 can be used for treating cancer or other diseases that cause disruptions of cell proliferation.
  • Pyrimidines and analogs are already described as active ingredients, such as, for example, the 2-anilino-pyrimidines as fungicides (DE-A-4029650) or substituted pyrimidine derivatives for treating neurological or neurodegenerative diseases (WO 99/19305).
  • pyrimidine derivatives As CDK-inhibitors, the most varied pyrimidine derivatives are described, for example bis(anilino)-pyrimidine derivatives (WO 00/12486), 2-amino-4-substituted pyrimidines (WO 01/14375), purines (WO 99/02162), 5-cyano-pyrimidines (WO 02/04429), anilinopyrimidines (WO 00/12486) and 2-hydroxy-3-N,N-dimethylaminopropoxy-pyrimidines (WO 00/39101).
  • bis(anilino)-pyrimidine derivatives WO 00/12486
  • 2-amino-4-substituted pyrimidines WO 01/14375
  • purines WO 99/02162
  • 5-cyano-pyrimidines WO 02/04429
  • anilinopyrimidines WO 00/12486
  • Protein ligands and receptor tyrosine kinases that specifically regulate endothelial cell function are substantially involved in physiological as well as in disease-related angiogenesis.
  • These ligand/receptor systems include the Vascular Endothelial Growth Factor (VEGF) and the Angiopoietin (Ang) families, and their receptors, the VEGF receptor family and the tyrosine kinase with immunoglobulin-like and epidermal growth factor homology domains (Tie) family.
  • VEGF Vascular Endothelial Growth Factor
  • Ang Angiopoietin
  • Tie immunoglobulin-like and epidermal growth factor homology domains
  • the VEGF receptor family includes Flt1 (VEGF-R1), Flk1/KDR (VEGF-R2), and Flt4 (VEGF-R3). These receptors are recognized by members of the VEGF-related growth factors in that the ligands of Flt1 are VEGF and placenta growth factor (PIGF), whereas Flk1/KDR binds VEGF, VEGF-C and VEGF-D, and the ligands of Flt4 are VEGF-C and VEGF-D (Nicosia, Am. J. Pathol. 153, 11-16, 1998).
  • the second family of endothelial cell specific receptor tyrosine kinases is represented by Tie1 and Tie2 (also kown as Tek).
  • Tie1 remains an orphan receptor
  • three secreted glycoprotein ligands of Tie2, Ang1 , Ang2, and Ang3/Ang4 have been discovered (Davis et al., Cell 87, 1161-1169, 1996; Maisonpierre et al., Science 277, 55-60, 1997; Valenzuela et al, Proc. Natl. Acad. Sci. USA 96, 1904-1909, 1999; patents: US 5,521 ,073; US 5,650,490; US 5,814,464).
  • Preferential inhibitors of the angiogenesis related kinases can be used for treating cancer or other diseases that are related to angiogenesis.
  • the object of this invention is to provide compounds that are inhibitors of cell cycle dependent kinases, in particular Chk, Akt, Pdk, CDK (cyclin dependent kinases) and/or angiogenesis related kinases, in particular VEGF-R (vascular endothelial growth factor receptor) kinases which have better properties than the inhibitors that are already known.
  • cell cycle dependent kinases in particular Chk, Akt, Pdk, CDK (cyclin dependent kinases) and/or angiogenesis related kinases, in particular VEGF-R (vascular endothelial growth factor receptor) kinases which have better properties than the inhibitors that are already known.
  • the substances that are described here are more effective, since they already inhibit in the nanomolar range and can be distinguished from other already known Cdk-inhibitors such as, e.g., olomoucine and roscovitine.
  • a or B in each case independently of one another represent cyano, halogen, hydrogen, hydroxy, aryl or the group -NO 2 , -NH 2 , - NR 3 R 4 , -d-e-alkyl-NR ⁇ 4 , -N(C 1 - 6 -hydroxyalkyl) 2l -NH-C(NH)-CH 3 , - NH(CO)-R 5 , -NHCOOR 6 , -NR 7 -(CO)-NR 8 R 9 , -NR 7 -(CS)-NR 8 R 9 , - COOR 5 , -CO-NR 8 R 9 , -CONH-C 1-6 -alkyl-COOH, -SO 2 -CH 3 , 4- bromo-1 -methyl- 1 H-pyrazolo-3yl or represent C ⁇ - 6 -alkyl optionally substituted in one or more places, the same way or differently with halogen, hydroxy, cyano or with the group
  • R 1 represents hydrogen, halogen, hydroxymethyl, C 1-6 -alkyl, cyano or the group -COOH, -COO-iso-propyl, -NO 2 , -NH-(CO)-(CH 2 ) 2 - COOH or -NH-(CO)-(CH 2 ) 2 -COO-C ⁇ -6 -alkyl, whereby the C 1-6 -alkyl can optionally be substituted in one or more places, in the same way or differently with halogen
  • R 2 represents hydrogen or the group -NH-(CO)-aryl or d- ⁇ -alkyl optionally substituted in one or more places, the same way or differently with cyano, hydroxy, aryl, heteroaryl, C 3-6 - heterocycloalkylring, which can optionally be interrupted with one or more nitrogen atoms, or substituted with the group -NR 8 R 9 , - NH-(CO)-NR 8 R 9 , -NH-(CO
  • R 6 represents C ⁇ - 6 -alkyl, C 2-6 -alkenyl or phenyl, whereby d- ⁇ -alkyl may optionally be substituted with C 3-6 - heterocycloalkylring that can optionally be interrupted with one or more nitrogen and/or oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, R 7 represents hydrogen or d- ⁇ -alkyl, R 8 or R 9 in each case independently of one another represent hydrogen, d -6 -alkyl, C 2-6 -alkenyl, C 3-6 -cycloalkyl, aryl or heteroaryl or the group whereby C ⁇ - 6 -alkyl, C 2-6 -alkenyl, C 3-6 -cycloalkyl, aryl or heteroaryl can optionally be substituted in one or more places, the same way or differently
  • R 10 represents -SO 2 -aryl, -SO 2 -heteroaryl or -SO 2 -NH 2 or -SO 2 -C ⁇ -6 - alkyl, whereby the aryl can be substituted with -d- 6 -alkyl, with the following provisos: whereby if X represents -NR 3 R 4 then R 2 does not represent a substituent, whereby if A and B represent hydrogen, X represents -NH- and R 2 represents d-e-alkyl, then R 1 represents -NH-(CO)-CH(NH 2 )-(CH2) 2 -COOH or -NH-
  • kinases which are involved in the regulation of the cell cycle, particulary Chks, Akt, Pdks and/or Cdks as well as angiogenesis related kinases, particulary VEGF-R kinases.
  • a compound refers to one or more of such compounds
  • the enzyme includes a particular enzyme as well as other family members and equivalents thereof as known to those skilled in the art.
  • Alkyl is defined in each case as a straight-chain or branched alkyl radical, such as, for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, hexyl, heptyl, octyl, nonyl and decyl.
  • Alkoxy is defined in each case as a straight-chain or branched alkoxy radical, such as, for example, methyloxy, ethyloxy, propyloxy, isopropyloxy, butyloxy, isobutyloxy, sec-butyloxy, tert-butyloxy, pentyloxy, isopentyloxy, or hexyloxy.
  • Hydroxy-Alkoxy is defined in each case as a straight-chain or branched alkoxy radical, such as, for example, methyloxy, ethyloxy, propyloxy, isopropyloxy, butyloxy, isobutyloxy, sec-butyloxy, tert-butyloxy, pentyloxy, isopentyloxy, or hexyloxy is substituted one or more times with hydroxy.
  • Alkylthio is defined in each case as a straight-chain or branched alkylthio radical, such as, for example, methylthio, ethylthio, propylthio, isopropylthio, butylthio, isobutylthio, sec-butylthio, tert-butylthio, pentylthio, isopentylthio or hexylthio.
  • Cycloalkyl is defined in general as monocyclic alkyl rings, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl or cyclodecyl, but also bicyclic rings or tricyclic rings such as, for example, norbornyl, adamantanyl, etc.
  • ring systems in which optionally one or more possible double bonds can be contained in the ring, are defined as, for example, cycloalkenyls, such as cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, or cycloheptenyl, whereby the linkage can be carried out both to the double bond and to the single bonds.
  • cycloalkenyls such as cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, or cycloheptenyl
  • R 3 and R 4 or R 8 and R 9 as defined in the claims in each case independently of one another, together form a C 3 -C ⁇ 0 -cycloalkyl ring, which optionally can be interrupted by one or more heteroatoms, such as nitrogen atoms, oxygen atoms and/or sulfur atoms, and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally one or more possible double bonds can be contained in the ring, however, the above-mentioned definitions are also intended to include heteroaryl radical or heterocycloalkyl and heterocycloalkenyl.
  • interrupted can mean either that the heteroatoms in addition to the carbon atoms form the ring or that the heteroatoms are substitutes for one or more carbon atoms.
  • Halogen is defined in each case as fluorine, chlorine, bromine or iodine.
  • alkenyl substituents in each case are straight-chain or branched, whereby, for example, the following radicals are meant: vinyl, propen-1-yl, propen-2-yl, but-1-en-1-yl, but-1-en-2-yl, but-2-en-1-yl, but-2-en-2-yl, 2-methyl-prop-2-en-1-yl, 2-methyl-prop-1-en-1-yl, but-1-en-3-yl, ethinyl, prop-1-in-1-yl, but-1-in-1-yl, but-2-in-1-yl, but-3-en-1-yl, and allyl.
  • Alkinyl is defined in each case as a straight-chain or branched alkinyl radical that contains 2-6, preferably 2-4 C atoms.
  • the following radicals can be mentioned: acetylene, propin-1-yl, propin-3-yl, but-1-in-1-yl, but-1-in-4-yl, but-2-in-1-yl, but-1-in-3-yl, etc.
  • the "aryl” radical in each case comprises 3-16 carbon atoms and in each case can be benzocondensed.
  • cyclopropenyl cyclopentadienyl
  • phenyl tropyl
  • cyclooctadienyl indenyl
  • naphthyl azulenyl
  • biphenyl fluorenyl, anthracenyl, etc.
  • heteroaryl radical in each case comprises 3-16 ring atoms, and instead of the carbon can contain one or more heteroatoms that are the same or different, such as oxygen, nitrogen or sulfur, in the ring, and can be monocyclic, bicyclic, or tricyclic and in addition in each case can be benzocondensed.
  • benzo derivatives thereof such as, e.g., quinolyl, isoquinolyl, etc., or azocinyl, indolizinyl, purinyl, etc. and benzo derivatives thereof; or quinolinyl, isoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, naphthyridinyl, pteridinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, xanthenyl, oxepinyl, etc.
  • Heterocycloalkyl stands for an alkyl ring that comprises 3- 6 carbon atoms, which can optionally be interrupted with one or more nitrogen and/or oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring.
  • interrupted can mean either that the heteroatoms in addition to the carbon atoms form the ring or that the heteroatoms are substitutes for one or more carbon atoms.
  • the heterocycloalkyl radical may be a monocyclic, or bicyclic ring system, which may include fused or bridged ring systems; and additionally the nitrogen or sulfur atoms in the heterocyclyl radical may be optionally oxidized; the nitrogen atom may be optionally quatemized; and the heterocyclyl radical may be aromatic or partially or fully saturated.
  • heterocycloalkyls there can be mentioned, e.g.: oxiranyl, oxethanyl, aziridinyl, azetidinyl, tetrahydrofuranyl, pyrrolidinyl, pyrrolidinonyl, dioxolanyl, imidazolidinyl, imidazolidinonyl, thiazolidiononyl, pyrazolidinyl, pyrazolidinonyl, dioxanyl, piperidinyl, piperidinonyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, trithianyl, quinuclidinyl, oxazolidinyl, oxazolidinonyl, hydantoin, pyran, thiin, dihydroacet, etc.
  • suitable conditions for carrying out a synthetic step are explicitly provided herein or may be discerned by reference to publications directed to methods used in synthetic organic chemistry.
  • the reference books and treatise set forth above that detail the synthesis of reactants useful in the preparation of compounds of the present invention, will also provide suitable conditions for carrying out a synthetic step according to the present invention.
  • "methods known to one of ordinary skill in the art” may be identified though various reference books and databases. Suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds of the present invention, or provide references to articles that describe the preparation, include for example, "Synthetic Organic Chemistry", John Wiley & Sons, Inc., New York; S. R.
  • Specific and analogous reactants may also be identified through the indices of known chemicals prepared by the Chemical Abstract Service of the American Chemical Society, which are available in most public and university libraries, as well as through on-line databases (the American Chemical Society, Washington, D.C. may be contacted for more details). Chemicals that are known but not commercially available in catalogs may be prepared by custom chemical synthesis houses, where many of the standard chemical supply houses (e.g., those listed above) provide custom synthesis services.
  • Solid compound and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • “Mammal” includes humans and domestic animals, such as cats, dogs, swine, cattle, sheep, goats, horses, rabbits, and the like.
  • Optional or “optionally” means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not.
  • optionally substituted aryl means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
  • “Pharmaceutically acceptable carrier, diluent or excipient” includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
  • “Pharmaceutically acceptable salt” includes both acid and base addition salts.
  • “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like.
  • “Pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2- dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like.
  • basic ion exchange resins such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-
  • organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline caffeine, N-methyl-glucamine, dimethyl-glucamine, ethyl-glucamine, lysine,
  • compounds which are "commercially available” may be obtained from standard commercial sources including Acros Organics (Pittsburgh PA), Aldrich Chemical (Milwaukee Wl, including Sigma Chemical and Fluka), Apin Chemicals Ltd. (Milton Park UK), Avocado Research (Lancashire U.K.), BDH Inc. (Toronto, Canada), Bionet (Cornwall, U.K.), Chemservice Inc. (West Chester PA), Crescent Chemical Co. (Hauppauge NY), Eastman Organic Chemicals, Eastman Kodak Company (Rochester NY), Fisher Scientific Co. (Pittsburgh PA), Fisons Chemicals (Leicestershire UK), Frontier Scientific (Logan UT), ICN Biomedicals, Inc.
  • suitable conditions for carrying out a synthetic step are explicitly provided herein or may be discerned by reference to publications directed to methods used in synthetic organic chemistry.
  • the reference books and treatise set forth above that detail the synthesis of reactants useful in the preparation of compounds of the present invention, will also provide suitable conditions for carrying out a synthetic step according to the present invention.
  • "methods known to one of ordinary skill in the art” may be identified though various reference books and databases. Suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds of the present invention, or provide references to articles that describe the preparation, include for example, "Synthetic Organic Chemistry", John Wiley & Sons, Inc., New York; S. R.
  • Specific and analogous reactants may also be identified through the indices of known chemicals prepared by the Chemical Abstract Service of the American Chemical Society, which are available in most public and university libraries, as well as through on-line databases (the American Chemical Society, Washington, D.C. may be contacted for more details). Chemicals that are known but not commercially available in catalogs may be prepared by custom chemical synthesis houses, where many of the standard chemical supply houses (e.g., those listed above) provide custom synthesis services.
  • Prodrugs is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of the invention.
  • prodrug refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable.
  • a prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound of the invention.
  • Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood.
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam).
  • prodrugs are provided in Higuchi, T., et al., "Pro-drugs as Novel Delivery Systems," A.C.S. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein.
  • prodrug is also meant to include any covalently bonded carriers which release the active compound of the invention in vivo when such prodrug is administered to a mammalian subject.
  • Prodrugs of a compound of the invention may be prepared by modifying functional groups present in the compound of the invention in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound of the invention.
  • Prodrugs include compounds of the invention wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively.
  • Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups in the compounds of the invention and the like.
  • “Therapeutically effective amount” refers to that amount of a compound of formula (I) which, when administered to a mammal, preferably a human, is sufficient to effect treatment, as defined below, for a disease-state alleviated by the inhibition of AKT-, PDK-, CHK-, CDK- or VEGF-R- acitivity, such as cancer.
  • the amount of a compound of formula (I) which constitutes a "therapeutically effective amount” will vary depending on the compound, the condition and its severity, and the age of the mammal to be treated, but can be determined routinely by one of ordinary skill in the art having regard to his own knowledge and to this disclosure.
  • Treating covers the treatment of disease-states alleviated by the inhibition of AKT-, PDK-, CHK-, CDK- or VEGF-R- activity, such as cancer, as disclosed herein, in a mammal, preferably a human, and includes:
  • the compounds of formula (I), or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or ( )- for amino acids.
  • the present invention is meant to include all such possible isomers, as well as, their racemic and optically pure forms.
  • Optically active (+) and (-), (f?)- and (S)-, or (D)- and ( )- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, such as reverse phase HPLC.
  • the formulae described herein contain olefinic double bonds or other centers of geometric asymmetry, unless specified otherwise, it is intended that the formulae include both E and Z geometric isomers, as well as all tautomeric forms.
  • all compound names herein, unless specified otherwise are intended to include all single enantiomers, diastereomers, and mixtures thereof, as well as racemic and non-racemic mixtures.
  • a or B in each case independently of one another represent cyano, halogen, hydrogen, hydroxy, tetrazolyl or the group -NH 2 , -NR 3 R 4 ,
  • R 1 represents cyano, hydrogen, halogen or C ⁇ -6 -alkyl, whereby the Ci.
  • 6 -alkyl can optionally be substituted in one or more places, in the same way or differently with halogen
  • R 2 represents hydrogen or the group -NH-(CO)-aryl or -C 1-6 -alkyl optionally substituted in one or more places, the same way or differently with cyano, hydroxy, aryl , heteroaryl, C 3-6 - heterocycloalkylring which can be optionally be interrupted in one or more places with one or more nitrogen atoms, or substituted with the group -NR 8 R 9 , -NH-(CO)-NR 8 R 9 , -NH-(CO)-S-C 1-6 -alkyl, - NH-(CS)-NR 8 R 9 , -NH(CO)-R 5 , -NH(CO)-OR 5 , -(CO)-NH-NH 2 , - (CO)-NH-CH 2 -(CO)-NH 2 , -(CO)-NH-C 1-6 -alkyl , -COOH whereby the aryl or the heteroaryl can optionally be substituted in one or more places,
  • R 3 or R 4 in each case independently of one another represent hydrogen, C ⁇ -6-alkyl optionally substituted in one or more places, the same way or differently with hydroxy, phenyl or hydroxyphenyl, or
  • R 3 and R 4 together form a C 3 -6-heterocycloalkylring containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C 3 -6- heterocycloalkylring can optionally be substituted with d -6 -alkyl, C 1-6 -alkyl-COOH or C 1-6 -alkyl-NH2, R 5 represents hydrogen, d -6 -alkyl, C ⁇ - 6 -alkoxy, C 2 - 6 -alkenyl, C 3-6 - cycloalkylring, heteroaryl , the group -(CO)-NH 2 or C 3 - 6 - heterocycloalkylring that can optionally be interrupted with one or more nitrogen and/or oxygen and/or sulfur atoms and/or can be interrupted by one or
  • R 8 or R 9 in each case independently of one another represent hydrogen
  • COOH, -NO 2 , or a C 3-6 -heterocycloalkylring can optionally be interrupted with one or more nitrogen and/or oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring or R 8 and R 9 together form a C 3-6 -heterocycloalkylring containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C 3-6 - heterocycloalkylring can optionally be substituted in one or more places, the same way or differently with hydroxy, hydroxy-C 1-6 -alkyl or the group -NR 8 R 9 , -NH(CO)-R 5 or -COOH and
  • R 10 represents -SO 2 -NH 2 , -SO 2 -C 1-6 -alkyl, -SO 2 -aryl, or -SO 2 - heteroaryl, whereby the aryl can be substituted with -d -6 -alkyl, as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
  • a or B in each case independently of one another represent hydrogen, tetrazolyl or the group -N(CH 3 ) 2 , -NH-(CO)-pyrrolidinyl, -NH-(CO)- pentyl, -NH-(CO)-hexyl, -NH-(CO)-hexyl-NH 2 , -NH-(CO)-C 3 H 7 , - NH-(CO)-CH 2 -phenyl, -NH-(CO)-CH 2 -NH 2 , -NH-(CO)-C 2 H 4 -NH 2 , - NH-(CO)-CH(NH 2 )-CH 3 , -NH-(CO)-CH(NH 2 )-hydroxyphenyl, -NH-NH-
  • R 1 represents halogen
  • R 2 represents hydrogen or the group -NH-(CO)-phenyl or -C 2 H -, -C 3 H 6 - both can optionally be substituted in one or more places, the same way or differently with cyano, hydroxy, phenyl, naphthyl, imidazolyl, thiazolyl, pyridyl, 2-oxazolinyl, piperidinyl, - NH 2 , -NH-CH 2 -thienyl, -NH-pyridinyl-NO 2 , -NH-thiazolyl, -SO 2 - thienyl, -SO 2 -NH 2 , -SO 2 -CH 3 , -SO 2 -C 3 H 7 , pyrrolidinonyl substituted with -COOH, -NH-(CO)-NH-thienyl, -NH-(CO)-NH-phenyl, -NH- (CO)-NH- C 2 H 5 ,
  • a or B in each case independently of one another represent hydrogen or the group -NH-(CO)-pyrrolidinyl, -NH-(CO)-piperidinyl, -NH-(CO)- morpholinyl, -NH-(CO)-hexyl-NH2, -NH-(CO)-CH(NH2)- hydroxyphenyl, -NH-(CO)-CH(NH2)-CH2-hydroxyphenyl, hydantoin optionally substituted with -CH3, X represents or the group -NH-,
  • R 1 represents halogen
  • R 2 represents hydrogen, -C 2 H 4 -imidazolyl or -C 3 H wich can optionally be substituted in one or more places, the same way or differently with the group -NH-CH 2 -thienyl, -NH-(CO)-C 2 H 5 , -NH-(CO)- C(CH 3 ) 3 ,
  • a or B in each case independently of one another represent hydrogen or the group -NO 2 , -NH 2 , -NR 3 R 4 , -N(C 1-6 -hydroxyalkyl) 2 , -NH(CO)- R 5 , -NHCOOR 6 , -NR 7 -(CO)-NR 8 R 9 , -NR 7 -(CS)-NR 8 R 9 , -COOR 5 , - CO-NR 8 R 9 , -SO 2 -CH 3 , 4-bromo-1-methyl-1H-pyrazolo-3yl or d-e-alkyl optionally substituted in one or more places, the same way or differently with cyano, halogen, hydroxy or the group -NH 2 , -NH-(CO)-R 5 , -SO 2 -NHR 3 , -COOR 5 , -CONR 8 R 9 , -O-(CO)-R 5 , -O- (CO)-C ⁇ -6
  • R 1 represents hydrogen, halogen, hydroxymethyl or the group -
  • R 2 represents C ⁇ -6-alkyl optionally substituted in one or more places, the same way or differently with hydroxy, imidazolyl or the group - NH 2 , -NH-(CO)O-CH 2 -phenyl, -NH-(CO)H, -NH-(CO)-phenyl, -NH- (CO)-CH 2 -O-phenyl, -NH-(CO)-CH 2 -phenyl, -NH-(CO)- CH(NH 2 )CH 2 -phenyl, -NH-(CO)-CH 2 -CH(CH 3 )-phenyl, -NH-(CO)- CH(NH2)-(CH2) 2 -COOH,
  • phenyl can optionally be substituted in one or more places, the same or differently with halogen, d -6 -alkyl or -(CO)- C(CH 2 )-C 2 H 5 , or represents C 3 -alkinyl,
  • R 3 or R 4 in each case independently of one another represent hydrogen or d- ⁇ -alkyl optionally substituted in one or more places, the same way or differently with hydroxy, phenyl or hydroxyphenyl, or R 3 and R 4 together form a C 3 - 6 -heterocycloalkylring containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupoted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C 3-6 - heterocycloalkylring can optionally be substituted with C 1-6 -alkyl, d- 6 -alkyl-COOH or C ⁇ -6 -alkyl-NH2, R 5 represents C ⁇ - 6 -alkyl, C 2-6 -alkenyl, C 3-6 -cycloalkyl or phenyl each can optionally be substituted in one or more places
  • R 7 represents hydrogen or d- ⁇ -alkyl
  • R 8 or R 9 in each case independently of one another represent hydrogen, d- 6 -alkyl, C 2 . 6 -alkenyl, C 3-6 -cycloalkyl, aryl or phenyl, whereby aryl or phenyl can optionally be substituted in one or more places, the same way or differently with hydroxy or the group -NO 2 or -N(C ⁇ - 6 - alkyl) 2 or R 8 and R 9 together form a C 3 .
  • 6 -heterocycloalkylring containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C 3-6 - heterocycloalkylring can optionally be substituted with the group - NH 2 , as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
  • a or B in each case independently of one another represent hydrogen or the group -NH-C 2 H 4 -OH, -NH-CH 2 -hydroxyphenyl, -NH-(CO)- pyrrolidinyl, -NH-(CO)-CH(NH 2 )-CH 2 -phenyl, -NH-(CO)-pentyl-NH 2 , -NH-(CO)-hexyl-NH 2 , -NH-(CO)-CH 2 -NH 2 , -NH-(CO)-CH(NH 2 )- hydroxyphenyl, -NH-(CO)-CH 2 -hydroxyphenyl, -NH-(CO)-CH 2 -hydroxyphenyl, -NH-(CO)-CH 2 - methylphenyl, -NH-(CO)-C 2 H 4 -dihydroxyphenyl, -NH-(CO)- CH(OH)-phenyl, -NH-(CO)-CH(NH 2 )
  • pyrrolidinyl can optionally be substituted with hydroxy or the group -NH 2
  • X represents an oxygen atom or the group -NH-
  • R 1 represents halogen or hydroxymethyl
  • R 2 represents -C 2 H 5 optionally substituted in one or more places, the same way or differently with hydroxy, imidazolyl or represents -C 3 H 7 or -C 4 H 8 optionally substituted in one or more places, the same way or differently with the group -NH 2 , -NH- (CO)-CH(NH 2 )-C 2 H 4 -COOH, -NH-(CO)-phenyl, -NH-(CO)-CH 2 - phenyl, -NH-(CO)-CH 2 -CH(CH 3 )-phenyl, -NH-(CO)-CH 2 -O-phenyl, -NH-(CO)O-CH 2 -phenyl, -NH-(CO)-CH(NH 2 )CH 2 -phenyl, -NH-(CO)-CH(NH 2 )CH 2 -phenyl,
  • phenyl can optionally be substituted in one or more places, the same or differently with halogen, -CH 3 or -(CO)- C(CH 2 )(C 2 H 5 ), or represents C 3 -alkinyl, as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
  • X represents the group -NH-
  • R 1 represents halogen and R 2 represents -CH 2 -dihydroxyphenyl, -C 2 H -imidazolyl, or -C 3 H optionally substituted in one or more places, the same way or differently with
  • compounds of the following structure are inhibitors of kinases, particularly AKT, PDK, Chk, CDK and/ or VEGF-R kinases:
  • compositions comprising as an active ingredient at least one compound of general formula (I) or compounds disclosed hereinbefore in an therapeutically effective amount for the prevention or treatment of a disorder caused by, associated with or accompanied by disruptions of cell proliferation and/or angiogenesis together with an pharmaceutically acceptable carrier, diluent or excipient.
  • a further object of the invention are use of a compound of general formula (I) or compounds disclosed hereinbefore for the manufacture of a medicament for the prevention or treatment of a disorder caused by, associated with or accompanied by any abnormal kinase activity selected from Chk, Akt, Pdk, Cdk and/or VEGF-R activity as well as combinations thereof.
  • kinase is selected from PDK1 , Akt1 , Akt2 and/or Akt3, particularly, wherein the kinase is selected from PDK1 , Akt1 , Akt2 and/or Akt3 in combination with VEGF-R or wherein the kinase is selected from Chk1 and/or Chk2.
  • Another objective of this invention is a method of treating a mammal having a disease-state alleviated by the inhibition of Akt, Pdk, chk and/or VEGF-R activity, wherein the method comprises administering to a mammal a therapeutically effective amount of a compound of general formula (I) or a compound disclosed hereinbefore.
  • the method is objective wherein the mammal is a human.
  • ..Disorders and/or additionallydisease state, in the meaning of this invention are selected from cancer, angiofribroma, arthritis, eye diseases, auto-immune diseases, chemotherapy agent-induced alopecia and mucositis, Crohn-disease, endometriosis, fibrotic diseases, hemangioma, cardiovaskular diseases, infectious diseases, nephrological diseases, chronic und acute neurodegenerative diseases, like disruptions of nerval tissue, viral infections, to prevent restenosis of vessels, for preventing the formation of scars, preventing or treating keratoma seniles and contact dermatitis, wherein cancer stands for solide tumours, tumour- or metastasis growth, Kaposis
  • fibrotic diseases stand for cirrhosis of the liver, mesangial cell proliferative diseases, arteriosklerosis, infectiouse diseases stand for diseases that are caused by unicellular parasites, cardiovascular diseases stand for stenosis, like stent induced restenosis, arteriosklerosis and restenosis, nephrological diseases stand for glomerulonephritis, diabetic nephropaty, malignant nephrosklerosis, thrombic mikroangiopathis syndrome, transplant rejections and glomerulopathy, chronic neurodegenerative diseases stand for Huntington's disease, amyotrophic lateralsklerosis, Parkinsons disease, AIDS
  • the compounds according to the invention essentially inhibit on the one hand cell- cycle-associated kinases, particularly serin/threonine kinases, more particularly cyclin-dependent kinases (Cdks), Chks, Akts and/or Pdks or VEGF-R kinases.
  • Cdks cyclin-dependent kinases
  • Chks e.g. Chk1 and/or Chk2
  • Akts e.g. Akt1 , Akt2 and/or Akt3 and/or Pdks, e.g. Pdk1.
  • the compounds according to this invention essentially inhibit angiogenesis related kinases, particularly tyrosine kinases, more particularly VEGF-R kinases.
  • the compounds of general formula (I) according to claims 2 to 5 show a preferentiality towards Akts, e.g. Akt1 , Akt2 and/or Akt3 and/or Pdks, e.g. Pdk1; the compounds of general formula (I) according to claims 6 to 8 show a preferentiality towards Chks, e.g.
  • Chk1 and/or Chk2 and the compounds of general formula (I) according to claims 9 and 10 show preferentiality towards Akts and VEGF-R kinases upon which is based their action, for example, against cancer, angiofribroma, arthritis, eye diseases, auto-immune diseases, chemotherapy agent-induced alopecia and mucositis, Crohn-disease, endometriosis, fibrotic diseases, hemangioma, cardiovaskular diseases, infectious diseases, nephrological diseases, chronic und acute neurodegenerative diseases, like disruptions of nerval tissue, viral infections, to prevent restenosis of vessels, for preventing the formation of scars, preventing or treating keratoma seniles and contact dermatitis.
  • Compounds of general formula (I) according to claims 9 and 10 show the advantage in the treatment of disorders to have an inhibiting effect of two ways, in particular the cell cycle inhibition and the angiogenesis inhibition due to the preferential inhibition of AKT and VEGF compounds.
  • the eukaryotic cell division ensures the duplication of the genome and its distribution to the daughter cells by passing through a coordinated and regulated sequence of events.
  • the cell cycle is divided into four successive phases: the G1 phase represents the time before the DNA replication, in which the cell grows and is sensitive to external stimuli. In the S phase, the cell replicates its DNA, and in the G2 phase, preparations are made for entry into mitosis. In mitosis (M phase), the replicated DNA separates, and cell division is completed.
  • the loss of the regulation of the cell cycle and the loss of function of the control points are characteristics of tumor cells.
  • the cell cycle is activated by a number of viruses, both by transforming viruses as well as by non-transforming viruses, to make possible the replication of viruses in the host cell.
  • the false entry into the cell cycle of normally post-mitotic cells is associated with various neurodegenerative diseases.
  • the mechanisms of the cell cycle regulation, their changes in diseases and a number of approaches to develop inhibitors of the cell cycle progression and especially the CDKs were already described in a detailed summary in several publications (Sielecki, T. M. et al. (2000). Cyclin-Dependent Kinase Inhibitors: Useful Targets in Cell Cycle Regulation. J. Med. Chem. 43, 1-18; Fry, D. W. & Garrett, M.
  • VEGF-C Consistent with the lymphatic expression of Flt4 in adults overexpression of VEGF-C in the skin of transgenic mice resulted in lymphatic, but not vascular, endothelial proliferation and vessel enlargement (Jeltsch et al., Science 276, 1423-1425, 1997). Moreover, VEGF-C was reported to induce neovascularization in mouse cornea and chicken embryo chorioallantoic membrane models of angiogenesis (Cao et al., Proc. Natl. Acad. Sci. USA 95, 14389-14394, 1998).
  • VEGF vascular endothelial growth factor
  • VEGF- neutralizing antibodies Interference with the VEGF/VEGF receptor system by means of VEGF- neutralizing antibodies (Kim et al., Nature 362, 841-844, 1993), retroviral expression of dominant negative VEGF receptor variants (Millauer et al., Nature 367, 576-579, 1994), recombinant VEGF-neutralizing receptor variants (Goldman et al., Proc. Natl. Acad. Sci. USA 95, 8795-8800, 1998), or small molecule inhibitors of VEGF receptor tyrosine kinase (Fong et al., Cancer Res. 59, 99-106, 1999; Wedge et al., Cancer Res. 60, 970-975, 2000; Wood et al.
  • a pharmaceutical preparation which in addition to the active ingredient for enteral or parenteral administration contains suitable pharmaceutical, organic or inorganic inert carrier materials, such as, for example, water, gelatin, gum arabic, lactose, starch, magnesium stearate, talc, vegetable oils, polyalkylene glycols, etc.
  • suitable pharmaceutical, organic or inorganic inert carrier materials such as, for example, water, gelatin, gum arabic, lactose, starch, magnesium stearate, talc, vegetable oils, polyalkylene glycols, etc.
  • the pharmaceutical preparations can be present in solid form, for example as tablets, coated tablets, suppositories, or capsules, or in liquid form, for example as solutions, suspensions, or emulsions.
  • adjuvants such as preservatives, stabilizers, wetting agents or emulsifiers; salts for changing the osmotic pressure or buffers.
  • injection solutions or suspensions especially aqueous solutions of active compounds in polyhydroxy-ethoxylated castor oil
  • carrier systems surface-active adjuvants such as salts of gallic acids or animal or plant phospholipids, as well as mixtures thereof and liposomes or ingredients thereof can also be used.
  • tablets, coated tablets, pills or capsules with talcum and/or hydrocarbon carriers or binders such as, for example, lactose, maize or potato starch
  • talcum and/or hydrocarbon carriers or binders such as, for example, lactose, maize or potato starch
  • the oral application can also be in a liquid form, such as, for example, as a juice, to which optionally a sweetener is added.
  • the dosage of the active ingredients can vary depending on the method of administration, age and weight of the patient, type and severity of the disease to be treated and similar factors.
  • the daily dose is 0.5-1000 mg, preferably 50-200 mg, whereby the dose can be given as a single dose to be administered once or divided into two or more daily doses.
  • the isomer mixtures can be separated into the enantiomers or E/Z isomers according to commonly used methods, such as, for example, crystallization, chromatography or salt formation.
  • the production of the salts is carried out in the usual way by a solution of the compound of formulae l-VII being mixed with the equivalent amount of or excess base or acid, which optionally is in solution, and the precipitate being separated or the solution being worked up in the usual way.
  • Inhibition of Pdk/Akt activity is carried out in the usual way by a solution of the compound of formulae l-VII being mixed with the equivalent amount of or excess base or acid, which optionally is in solution, and the precipitate being separated or the solution being worked up in the usual way.
  • PDK-1 phosphoinositide-dependent kinase-1
  • the compounds therefore, can be blocking the assay by inhibiting PDK-1 enzyme activity, AKT enzyme activity, or the activation of AKT by PDK-1.
  • These compounds are expected to be therapeutically useful in cancer by inhibiting processes critical for tumor progression, including cell proliferation, survival, and tumor angiogenesis (Testa and Bellacosa 2001; Vivanco and Sawyers 2002).
  • compounds blocking block colony formation and/or growth of PC-3 prostate and MDA-468 breast cancer cells in soft agar, which is an in vitro measure of potential anti-tumor activity. Furthermore, the compounds described herein are expected to sensitize tumors to the effects of other chemotherapeutic agents and radiation (Page, Lin et al. 2000; Brognard, Clark et al. 2001).
  • PDK-1 is a Ser/Thr kinase that functions to phosphorylate and activate other Ser/Thr kinases in the AGC kinase family (Vanhaesebroeck and Alessi 2000).
  • the best-characterized substrate of PDK-1 is the intracellular Serine/Threonine kinase AKT, whose expression and/or activity is elevated in many cancers.
  • AKT glucocordicoid regulated kinase
  • SGK glucocordicoid regulated kinase
  • ErbB2 and IGF 1 -receptor ErbB2 and IGF 1 -receptor
  • oncogenes e.g, Ras, BCR-abl, and Src
  • Other potential substrates of PDK-1 include p70 S6 kinase, p90 S6 kinase, protein kinase C, cAMP-dependent protein kinase (PKA), PRK1 , Protein kinase G and serum and glucocorticoid regulated kinase (SGK).
  • PDK-1 -mediated phosphorylation of AKT which is largely in an inactive form in unstimulated cells, converts the enzyme to a catalytically active form. This occurs through the phosphorylation of the activation loop domain of AKT e.g., at Threonine-309 in AKT2 and Theonine-308 in AKT1. Phosphorylation of a homologous domain in many kinases is known to regulate their kinase activity.
  • One stimulus for PDK-1 mediated phosphorylation of AKT is the association PI-3 kinase products (3,4,5)PIP 3 or (3,4)PIP 2 with the pleckstrin homology (PH) domain of AKT.
  • AKT displays low, basal levels of activation in normal, unstimulated cells
  • AKT often becomes constitutively activated in tumor cells. This occurs through the up-regulation of a variety of different signaling molecules or the presence of oncogenenic mutations commonly found in cancer cells that can promote the activation of AKT, such as PI-3 kinase, growth factor receptors (e.g., EGFR family members), Ras, Src, and BCR-ABL activation.
  • Loss of the tumor suppressor PTEN is another means of greatly increasing AKT activity in cancer cells (Besson, Robbins et al. 1999).
  • PTEN mutation or down regulation of PTEN protein is found in a large number of tumors and cancer cell lines.
  • PTEN is a phosphatase that removes the D-3 phosphate from the products of PI-3 kinase such as phosphatidylinositol 3,4,5-trisphosphate and phosphatidylinositol 3,4-bisphosphate (Myers, Pass et al. 1998; Stambolic, Suzuki et al. 1998). Loss of PTEN, therefore, has the effect of increasing products of PI-3 kinase and promoting constitutive activation of AKT. Cancers with highly up-regulated levels of AKT may be especially sensitive to the effects of PDK-1 /AKT pathway inhibitors.
  • Downstream substrates of PDK-1 and/or AKT are associated with a number of cell responses including proliferation, metabolism and cell survival (Testa and Bellacosa 2001; Vivanco and Sawyers 2002).
  • Examples of signaling molecules downstream from PDK-1 or AKT involved in these pathways include BAD, p70 S6 kinase, p21(Waf-1/Cip-1), Forkhead transcription factors, p27(kip-1), GSK-3-alpha/beta, TSC2 (tuberin), and ecNOS.
  • the survival function of AKT is particularly well-characterized cellular activity of AKT (Datta, Brunet et al. 1999).
  • AKT functions to suppress apoptosis induced by a variety of agents, including UV radiation, chemotherateutic drugs, TFG-beta, withdrawal of survival factors, overexpression of oncogenes such as c-myc and detachment of cells from the extracellular matrix.
  • apoptosis The ability to escape cell death, also termed apoptosis, is critical characteristic of tumor cells allowing their uncontrolled growth and invasive behavior.
  • One trigger for apoptosis is the perturbation of the normal growth regulation resulting from oncogenic mutations or inappropriate expression signaling molecules coupled to cell proliferation. Apoptotic pathways, therefore, provide a key means of protection from the development and progression of cancer.
  • Cancer cells can escape apoptotic death by selecting for activation of signaling molecules such as AKT that turn off apoptotic signals.
  • Some oncogenes, such as Ras which is activated in as many as 60% of human tumors, simultaneously promote uncontrolled growth and the activation of AKT.
  • Inhibition of AKT in HIH 3T3 cells prevents transformation of these cells through transfection with activated Ras. Furthermore, a number of studies have shown that combining expression an oncogene with an activated form of AKT greatly facilitates formation of tumors in vivo (e.g., (Holland, Celestino et al. 2000)). Inhibitors of PDK-1, by blocking activation of AKT, are a means of promoting apoptosis in tumors cells, especially, but not necessarily limited to those over-expressing AKT activity. By blocking cell survival mechanisms, the compounds described herein could also be useful to promote sensitivity of cancer cells to radiation therapy and to treatment with a variety of chemotherapeutic agents.
  • Inhibitors of the PDK-1 /AKT pathway are also expected to block cancer progression through inhibition of tumor-stimulated angiogenesis (Dimmeler and Zeiher 2000; Shiojima and Walsh 2002).
  • AKT has been shown to regulate a number of responses critical for the process of angiogeneisis, including endothelial cell migration, proliferation and survival during new vessel formation, ecNOS regulation, response of endothelial cells to growth factors (including IGF-1 , agniopoetin-1 and VEGF) and the regulation of hypoxia-inducible factor-1 (HIF-l)-alpha levels.
  • Inhibition of the cell cycle and growth of tumor cells is yet another expected effect of compounds that block PDK-1 and/or AKT.
  • Inhibition of PDK-1 and/or AKT activity has been shown to regulate growth of cancer cells in a number of studies. These effects may occur through PDK-1 or AKT-mediated regulation of a number of different signaling pathways important in growth regulation.
  • AKT has been shown to block nuclear localization and/or expression of the cyclin-dependent kinase inhibitors, p21(Waf-1/Cip-1) and p27(kip-1). Compounds blocking these effects would be expected to reduce the activity of cyclin-dependent kinases, blocking progression through the cell cycle and reducing tumor cell growth.
  • AKT was found to inhibit Myt1 , thereby acting as an initiator of mitosis in oocytes fronm the starfish Asterina pectinfera. Furthermore, PDK-1 and/or AKT regulate the expression of proteins important for cell growth through its regulation of mTOR, p70 S6 kinase and eukaryotic initiation factor 4E binding protein 1 (4E-BP1). While the mechanism of this regulation is not firmly established, it has been shown that AKT phosphorylations and reduces expression of TSC2, thereby relieving TSC-2 mediated suppression of mTOR activity.
  • the compounds described which block PDK-1 mediated activation of AKT or PDK-1 directly may be useful therapeutic agents in cancer by blocking a number of processes required for tumor progression, including growth, tumor cell survival, and recruitment of new blood vessels.
  • the compounds described may also enhance the anti-tumor effects of radiation or other chemotherepeutic drugs.
  • the compounds may also be useful for the treatment of tuberous sclerosis.
  • the compounds described could be useful modulators of the immune response (Cantrell 2002) and for the treatment of autoimmune diseases such as rheumatoid arthritis and MS.
  • Prostate cancer cells PC-3) and breast cancer cells (MDA-468) were obtained from the ATCC (Manassas, VA).
  • Mammalian protein extraction reagent MPER
  • Halt protease inhibitor cocktail BCA protein reagent
  • Supersignal Western Chemiluminescent reagent were obtained from Pierce Chemical Co. (Rockford, IL).
  • 10% Tris-Glycine gels 1.0mm, 15-well
  • nitrocellulose 0.2 micron
  • Agar agar was purchased from EM Science.
  • Polyclonal antibodies raised against phospho-AKT (Thr308, #9275), phospho-AKT (Ser473, #9271), phospho-S6-kinase (Thr389, #9205), and anti-rabbit IgG-HRP conjugate were obtained from Cell Signaling Technologies (Beverly, MA). Nitroblue tetrazolium reagent and staurosporine were purchased from Sigma Chemical Co. (St. Louis, MO). LY294002 was purchased from Cayman Chemicals (Ann Arbor, Ml). All other materials were of reagent-grade quality.
  • PC-3 cells were grown in F12K medium, supplemented with 7% (v/v) fetal calf serum (fcs) and 2mM glutamine.
  • MDA-468 cells were grown in MEM-alpha, supplemented with 10% (v/v) fcs, 2mM glutamine, 1mM sodium pyruvate, 0.1 mM non-essential amino acids, 10mM Hepes, and 1 ⁇ g/ml insulin. All cell lines were incubated in a 37DC humidified incubator, with a 5% CO 2 atmosphere.
  • PC-3 cells were seeded into 24-well plates (Corning Costar) at 100-120,000 cells per well and allowed to grow overnight to 90% confluence. On the next day, the cells were washed once with 1.5ml PBS, and the medium replaced with low serum (0.1% fcs) containing growth medium (starvation medium). After a second overnight incubation, the medium was replaced with 0.5ml/well of starvation medium. Some assays were also conducted in normal growth medium (7% fcs, PC-3, or 10% fcs, MDA-468).
  • DMSO vehicle control
  • drug a final DMSO concentration of 1% v/v (a 5 ⁇ l addition per 0.5ml medium)
  • the incubations were terminated by aspiration of the medium, washing the wells with 1.0ml PBS, and lysis in 0.1ml MPER reagent, supplemented with protease inhibitors (Halt reagent) and phosphatase inhibitors (1mM NaF, 1mM sodium vanadate).
  • Halt reagent protease inhibitors
  • phosphatase inhibitors 1mM NaF, 1mM sodium vanadate
  • lysates were combined with Laemmli SDS sample buffer, boiled, and loaded onto 10% Tris-Glylcine gels, normalizing for the amount of protein loaded in each lane. Electrophoresed gels were transferred onto nitrocellulose paper, blocked with 5% milk in Tris-buffered saline containing 0.1% Tween-20, and incubated overnight with the primary antibody (phospho-AKT-Thr308 @ 1 :667, phospho-AKT-Ser473 @ 1:1000, phospho-S6 kinase @ 1 :1000). Blots were washed three times with blocking buffer and incubated one hour with anti-rabbit IgG-HRP @ 1:2000. Washed blots were developed using the Supersignal Western Chemiluminescent detection system. Films were scanned using a Bio Rad CCD camera, and phospho-protein bands were quantitated using Bio Rad Quantity-One software.
  • Soft agar efficacy assays PC-3 and MDA-468 cells were grown in soft agar over a period of 2 weeks. Culture plates (Corning 35mm x 10mm) were prepared with a bottom layer of 0.5% agar in growth medium, 2ml/well. Cells were trypsinized, dispersed into single cells with a 21 -gauge needle, and seeded in a top layer of 0.3% agar/growth medium, 1.5ml/plate, containing 4500 cells per plate. On the following day, the first vehicle or drug treatment was added, in a volume of 1.0ml of 0.3% agar/growth medium, containing 1% DMSO. Drug concentrations were adjusted to reflect the total volume of agar in the plates.
  • the cells were allowed to grow for seven days and treated a second time (adding an additional 1 ml of 0.3% agar). Colonies were visually inspected for growth and viability every few days. On day 12-14, nitroblue tetrazolium (0.5 mg/ml PBS) was added, 0.3 ml per plate, and the viable colonies were allowed to develop color for 1-2 days. Plates were scanned using a Bio Rad CCD camera, and the colonies were quantitated for ony number, and for total stained area, using ImagePro software.
  • pHisAKT2 was constructed by cloning AKT2 into pBlueBacHis2A (Invitrogen Corp.) through the BamH1 and Bgl2 restriction sites, forming a fusion protein behind a 38 amino acid N-terminal His tag sequence derived from the vector.
  • the new N-terminal sequence + first 10 residues of AKT2 is as follows: MPRGSHHHHHHGMASMTGGQQMGRDLYDDDDKDRWGSMNEVSVIKEG (AKT2 is underlined and is in bold His-6).
  • pHisPDK-1 was constructed by cloning PDK1 into pBlueBacHis2A (Invitrogen Co ⁇ .) at EcoR1 cloning site, forming a fusion protein behind an N-terminal His-tag (preceding sequence of ...ICSWYHGILDMARTTSQLYD.... (PDK1 sequence underlined).
  • the new N-terminal sequence + first 10 residues of PDK1 is as follows: MPRGSHHHHHHGMASMTGGQQMGRDLYDDDDKDRWGSELEICSWYHGILD MARTTSQLYD- (PDK1 is underlined and His-6 is in bold).
  • Recombinant baculovirus containing either His-tagged AKT2 or His-tagged PDK-1 cDNAs were prepared by the following method. pHisAKT2 or pHisPDK-1 were cotransfected with Bac-N-Blue (Invitrogen) viral DNA info SF-21 cells and after 3 - 4 days, viral supernatant were isolated and recombinant viruses were plaque purified. His-tagged AKT2 (HisAKT-V) or His-tagged PDK-1 (HisPDK-1-V) cDNA expressing clones were selected and expanded as a stock for use in the expression of recombinant proteins described below.
  • pHisAKT2 or pHisPDK-1 were cotransfected with Bac-N-Blue (Invitrogen) viral DNA info SF-21 cells and after 3 - 4 days, viral supernatant were isolated and recombinant viruses were plaque purified. His-tagged AKT2 (HisAKT-V) or His-tagged PDK-1 (HisPDK-1-
  • SF-21 insect cells were infected with recombinant viruses (i.e., either HisPDK-1-V or HisAKT2-V) and cells were harvested 3-4 days post infection and frozen.
  • recombinant viruses i.e., either HisPDK-1-V or HisAKT2-V
  • cell pellets were thawed, homogenized (in phosphate buffered saline (PBS), supplemented with 10% Triton X-100, 0.5 M NaCl, 2 g/l NaF, 2.5 ⁇ g/ml aprotinin, 5 ⁇ g/ml leupeptin, 1.25 ⁇ g/ml pepstatin, 0.1% beta-mecaptoethanol, and 1 mM vanidate, 10 mM imidizole and adjusted to pH 7.6) and were purified using two sequential rounds of Ni2+ affinity chromatography followed by gel filtration.
  • PBS phosphate buffered saline
  • Enzymes were frozen in small aliquots and stored at -80DC in 50 mM Tris-HCl, pH 7.5, 150 mM NaCl, pH 7.5, 0.1 mM EGTA, 0.1 mM EDTA, 0.2 ⁇ M benzamidine, 0.1% beta-mercaptoethanol and 0.25 M sucrose.
  • PDK-1 -dependent activation and subsequent enzymatic activity of AKT2 Purified human AKT2 activity was routinely measured in an assay in which the enzyme was first activated by PDK-1 in the presence of phosphatidylinositol-4,5-bisphosphate (PIP2). Once activated, AKT2-dependent phosphorylation of a peptide substrate was measured by scintillation proximity assay (SPA).
  • PIP2 phosphatidylinositol-4,5-bisphosphate
  • Phospholipid vesicles were prepared as follows: 2.2 mg each of phosphatidylcholine (Sigma Cat # P-1287) and phosphatidylserine (Sigma Cat #P-6641) were transferred to a borosilicate glass test tube and dried down under nitrogen. 1 mg of PIP 2 (Biomol Cat #PH-106) was suspended in 9.5 ml of 10 mM HEPES, pH 7.5 and transferred to the dried lipids. The tube was vortexed until a milky suspension was produced.
  • the tube was placed in a ice water-jacketed cup horn sonicator (Branson Instruments) and subjected to sonication for 20 min at medium power until a translucent phospholipid vesicle preparation was obtained. Aliquots of the vesicle suspension were frozen at -80DC until needed.
  • Assays were performed in 96-well polypropylene V-bottom plates. Incubations were carried out for 2 hr at room temperature.
  • the assay mixture contained in a volume of 60 ⁇ L: 15 mM MOPS, pH 7.2, 1 mg/ml bovine serum albumin, 18 mM betaglycerolphosphate, 0.7 mM dithiothreitol, 3 mM EGTA, 10 mM MgOAc, 7.5 (M ATP, 0.2 ⁇ Ci of [ ⁇ - 33 P]ATP, 7.5 ⁇ M biotinylated peptide substrate (biotin-ARRRDGGGAQPFRPRAATF), 0.5 ⁇ L of PIP 2 -containing phospholipid vesicles, 60 pg of purified recombinant human PDK-1 , and 172 ng of purified recombinant human AKT2.
  • Test compounds were added from stock solutions in DMSO. The final concentration of DMSO was 2.5%. Following incubation, 10 ⁇ L of the assay mixture was transferred to a 96-well clear-bottom polystyrene plate (Wallac Isoplate) containing 0.33 mg of streptavidin-coated SPA beads (Amersham Cat. # RPNQ0007) suspended in 200 ⁇ L of phosphate-buffered saline, pH 7.4, containing 50 mM EDTA and 0.1% Triton X-100. After brief shaking, the SPA beads were allowed to settle to the bottom of the plate overnight at room temperature.
  • Product formation measured in a Wallac MicroBeta scintillation counter, was proportional to the time of incubation and to the amount of PDK-1 and inactive AKT2 added.
  • PDK-1 was added at sub-optimal levels so that the assay could sensitively detect inhibitors of AKT2 activation as well as direct AKT2 kinase inhibitors.
  • the z'-factor for the assay was greater than 0.7.
  • Phosphorylation of the peptide substrate on the threonine residue was shown to be dependent upon activated AKT2 produced during the incubation. No phosphorylation was observed in the absence of ATP, Mg 2 +, PDK-1 , AKT2, or PIP 2 -containing vesicles. Phosphorylation was readily observed, however, upon addition of purified activated human AKT1 (purchased from Upstate Biotechnology), independent of the presence or absence of added PDK-1 or PIP 2 -containing vesicles.
  • Direct assay of PDK-1 activity Recombinant human PDK-1 activity was directly measured using a filter binding protocol. Incubations were performed at room temperature for 4 hr in a final volume of 60 ⁇ L containing: 50 mM Tris-HCl, pH 7.5, 0.1 mM EGTA, 0.1 mM EDTA, 0.1% beta-mercaptoethanol, 1 mg/ml bovine serum albumin, 10 mM MgOAc, 10 ⁇ M ATP, 0.2 ⁇ Ci of [ ⁇ - 33 P]ATP, 7.5 ⁇ M of substrate peptide (H 2 N-ARRRGVTTKTFCGT) and 60 ng of purified human PDK-1.
  • the enzymatic reaction was stopped by addition of 25 mM EDTA. A portion of the reaction mixture was spotted on Whatman P81 phosphocellulose paper. The filter paper was washed 3 times with 0.75% phosphoric acid to remove unreacted [ ⁇ - 33 P]ATP, and once with acetone. After drying, the filter-bound labeled peptide was quantitated using a Fuji Phosphoimager. Results
  • Akt/protein kinase B is constitutively active in non-small cell lung cancer cells and promotes cellular survival and resistance to chemotherapy and radiation. Cancer Res 61(10): 3986-97.
  • TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nat Cell Biol 12: 12.
  • the compounds of this invention inhibit the cell cycle checkpoint kinases which are essential for the cellular response to DNA damage and for the coordination of the cell cycle.
  • the DNA damage might be due to external or internal influence. These influences involve - without being limited to them - replication errors, DNA base damages, DNA strand breaks and the exposition to irradiation or cytotoxic chemicals.
  • the inhibition of one or more of the cell cycle checkpoint kinases is the basis for the effect of the compounds of this invention e.g. against cancer, like solid tumours or leukemia, against other hyperproliferative diseases, e.g. HIV and viral infections, like e.g. cytomegalus-infections, herpes and hepatitis B and C and HIV.
  • cancer like solid tumours or leukemia
  • hyperproliferative diseases e.g. HIV and viral infections, like e.g. cytomegalus-infections, herpes and hepatitis B and C and HIV.
  • the eukaryotic cell division cycle ensures the duplication of the genome and its correct distribution to the daughter cells by running through a coordinated and regulated sequence of events.
  • the cell cycle is divided in four successive phases: the G1 phase represents the time before the DNA replication, during which the cell is growing and susceptible for external stimuli.
  • the S-phase the cell replicates its DNA
  • the G2 phase the cell prepares for the entry into the mitosis.
  • the mitosis M-Phase
  • the replicated DNA is separated and the cell division is carried out.
  • the passage through the cycle is strictly regulated and controlled.
  • the enzymes needed for the progression through the cycle, the cyclin-dependent kinases, have to be activated at the right moment and have to be switched off as soon as the corresponding phase is finished.
  • Checkpoint systems arrest the progression through the cell cycle if DNA damage is detected, the DNA replication is not completed or the building of the spindel apparatus is not completed (Hartwell et al., 1989). They do this by influencing the generation, activation or inactivation of the cyclin-dependent kinases.
  • Checkpoints permit the cell to track the ordered course of the individual phases of the cell cycle. The most important checkpoints are at the transition from the G1 phase into the S phase and at the transition from the G2 phase into the M phase
  • the G1 checkpoint ensures that the cell does not start the DNA synthesis if it is not sufficiently nourished or if it does not correctly interact with other cells or with the substrate or if the DNA of the cell is not intact.
  • the G2/M checkpoint ensures that the DNA is completely replicated and the mitotic spindle is build up before the cell enters the mitosis.
  • the G1 checkpoint is controlled by the gene product of the tumour suppressor gene p53. p53 becomes activated after the detection of changes in the metabolism or the genomic integrity of the cell and p53 is able to initiate either a stop of the cell cycle program or apoptosis. For this the transcriptional activation of the expression CDK inhibiting protein p21 plays a crucial role.
  • a fundamental component of the G2/M checkpoint is the activation of the kinases ATM, Chk1 and Chk2 after a DNA damage and finally the phosphorylation and inactivation of the phosphatase Cdc25C. This results in a cell cycle arrest, as the inhibitory phosphorylation of the amino acids threonine-14 and tyrosine-15 of the cyclin dependent kinase 1 (CDK1) is not further removed by Cdc25C.
  • CDK1 cyclin dependent kinase 1
  • tumour cells which is essential for the G1 checkpoint, is the gene most often mutated in human tumours (about 50 %). In tumour cells expressing unmutated p53, it is often inactivated by an enhanced proteolytic degradation or the genes of other proteins involved in the G1 checkpoint are mutated or deregulated. Examples are the inactivation of the tumour suppressor genes Rb, p16 INK 4 and p19 ARF or the overexpression of the oncogenes HDM-2 and cyclin D (Levine, 1997).
  • tumour cells do not have a functional G1 checkpoint which enables the to accumulate further mutations and to escape from a DNA damage induced apoptosis.
  • This inactivation of the G1 checkpoint is an important factor for the genomic instability which drives the evolution of human tumours and crucially contributes to the resistance of tumour cells against chemotherapeutics and irradiation.
  • the inactivation of the G1 checkpoint enhances the dependence of the tumour cells on the second important barrier against the cell killing effect of DNA damages, the G2/M checkpoint, and makes the tumour cells especially vulnerable to an abrogation of the G2/M checkpoint (Hartwell und Kastan, 1994, O'Connor und Fan, 1996).
  • the cell cycle checkpoint kinase Chk1 is an important part of the G2/M checkpoint (Sanchez et al., 1997). Inactivation of Chk1 abrogates a DNA damage induced G2/M arrest and thereby leads to a preferred killing of the resulting checkpoint deficient cells (Takai et al., 2000, Koniaras et al., 2001 , Liu et al., 2000). The inactivation of Chk1 causes that Cdc25C stays active despite of the DNA damage and is able to activate Cdk1/CycB, the main effector of the entry into the mitosis.
  • Chk2 is also activated by DNA damage (Matsuoka et al. 1998, Chaturvedi et al., 1999) and activated Chk2 phosphorylates and thereby inactivates Cdc25C.
  • Cells without active Chk2 have a defect in their checkpoint response to DNA damage (Hirao et al., 2000).
  • the inactivation of Chk1 and Chk2 abrogates the G2/M arrest which is induced by damaged DNA and sensitises the resulting checkpoint deficient cells to the killing by DNA damaging events.
  • DNA damaging events can be the direct damage of the DNA by irradiation or chemotherapeutics, e.g. strandbreaks inducing compounds, DNA-alkylating compounds or topoisomerase inhibitors, the exertion of influence on the building of the mitotic spindle apparatus, hypoxic stress due to limited supply of the tumour with blood - e.g. induced by anti-angiogenic drugs - or also endogenous DNA damages resulting from the genomic instability inherent to cancer cells.
  • chemotherapeutics e.g. strandbreaks inducing compounds, DNA-alkylating compounds or topoisomerase inhibitors
  • hypoxic stress due to limited supply of the tumour with blood - e.g. induced by anti-angiogenic drugs - or also endogenous DNA damages resulting from the genomic instability inherent to cancer cells.
  • Recombinant Chk1-His 6 -fusion protein expressed in insect cells (Sf-9) and purified by Ni-NTA affinity chromatography was used as kinase.
  • commercially available GST-Chk1-fusion protein (Upstate Biotechnology, Dundee, Scotland) can be used.
  • substrate for the kinase reaction the biotinylated peptide biotin-Arg-Ser-Gly-Leu-Tyr-Arg-Ser-Pro-Ser-Met-Pro-Glu-Asn-Leu-Asn-Arg-Pro- Arg-OH was used which can be purchased e.g. from the company Biosyntan GmbH (Berlin-Buch, Germany).
  • Chk1 (200 ng/measurement point) was incubated for 60 min at 22 DC in the presence of different concentrations of test compounds (0 ⁇ M and concentrations in the range 0.001 - 30 ⁇ M) in 30 ⁇ l assay buffer [50 mM Hepes/NaOH pH7.5, 10 mM MgCI 2 , 1 mM MnCI 2 , 0.1 mM sodium ortho-vanadate, 1.0 mM dithiothreitol, 0.5 ⁇ M adenosine-tri-phosphate (ATP), 1.9 ⁇ M substrate peptide
  • EDTA/ATP-solution (20 mM EDTA, 50 ⁇ M ATP, 1 % (v/v) Triton X-100 in PBS).
  • Recombinant Chk2-His 6 -fusion protein expressed in insect cells (Sf-9) and purified by Ni-NTA affinity chromatography was used as kinase.
  • commercially available GST-Chk2-fusion protein (Upstate Biotechnology, Dundee, Scotland) can be used.
  • substrate for the kinase reaction the biotinylated peptide biotin-Arg-Ser-Gly-Leu-Tyr-Arg-Ser-Pro-Ser-Met-Pro-Glu-Asn-Leu-Asn Arg-Pro-Arg-OH was used which can be purchased e.g. from the company Biosyntan GmbH (Berlin-Buch, Germany).
  • Chk2 (400 ng/measurement point) was incubated for 60 min at 22DC in the presence of different concentrations of test compounds (0 ⁇ M and concentrations in the range 0.001 - 30 ⁇ M) in 30 ⁇ l assay buffer [50 mM Hepes/NaOH pH7,5, 10 mM MgCI 2 , 1 mM MnCI 2 , 0.1 mM sodium ortho-vanadate, 1.0 mM dithiothreitol, 1.5 ⁇ M adenosine-tri-phosphate (ATP), 8 ⁇ M substrate peptide (Biotin-Arg-Ser-Gly-Leu-Tyr-Arg-Ser- Pro-Ser-Met-Pro-Glu-Asn-Leu-Asn-Arg-Pro-Arg-OH), 15 nCi/measurement point 33 P-gamma ATP, 0.008% NP40, 1.5% (v/v) dimethylsulfoxide]. The reaction was stopped by the addition of 20 ⁇ l
  • PVT-SPA-beads (0.25 mg/measurement point, from Amersham Biotech) in an aqueous EDTA/ATP-solution (20 mM EDTA, 50 ⁇ M ATP, 1 % (v/v) Triton X-100 in PBS).
  • Human HeLa (ATCC CCL-2) cervix adenocarcinoma cells were plate out to a density of 3000 cells / cm 2 in DMEM medium containing 10% FCS in 6-well plates. After 48 h incubation the medium was exchange for DMEM medium supplemented with 10% FCS and 5 ⁇ g/ml bleomycine sulfate. After 18 h incubation the test compounds were added to final concentrations of 0.03 ⁇ M, 0.1 ⁇ M, 0.3 ⁇ M, 1 ⁇ M,3 ⁇ M, 10 ⁇ M, or 30 ⁇ M. After a further incubation of 24 h or 48 h the cells were collected by trypsinisation, permeablelised and fixed in 70 % ethanol .
  • the DNA was stained with propidium iodide and the cellular DNA-content was measured by a Fluorescence Activated Cell Scan (FACS). The portion of cells with a cellular DNA-content corresponding to the G2 and M phases of the cell cycle was evaluated to judge the effect of the test compound on the bleomycine induced G2/M arrest of the cells.
  • FACS Fluorescence Activated Cell Scan
  • the coding sequences were cloned by RT-PCR and nested PCR from commercially available polyA-RNA.
  • the primers used for this purpose were designed according to the sequence information in Genebank (AF 016582 for Chk1, AF086904 for Chk2).
  • 3'-primers were used, which removed the stop codon at the end of the coding sequence of Chk1 and Chk2 by mutation. Additional restriction sites were added to the primers (EcoRI-sites for the 5'-primers and Hindlll-sites for the 3'-primers).
  • the cDNAs were cloned into the vector pT7-Blue T (Novagen). To introduce the His 6 -sequence at the C-terminus of Chk1 and Chk2 EcoRI/Hindlll fragments from these pT7-Blue plasmids were cloned into the bacterial expression vector pET23a. From these pET23a-Chk1 und pET23a-Chk1 vectors DNA fragments coding for Chk1-His6 or Chk2-His 6 were excised and inserted into the baculovirus-transfer-vector pVL1392. The generated vectors were transfected into Sf-9 cells with AcNPV baculovirus genomic DNA (BaculoGold Transfection Kit, Pharmingen). The viruses produced by this procedure were plaque-purified and amplified for further infections.
  • Recombinant Chkl-His ⁇ -fusion protein and recombinant Chk2-His 6 -fusion protein were produced in Sf-9-cells.
  • MOI Multiplicity of infectivity
  • the fusion proteins were purified from the supernatant by Ni-NTA affinity chromatography (Superflow from QIAGEN, Hilden, Germany) and dialysed into 50 mM Tris/HCI buffer (pH 7.5) containing 150 mM NaCl and 2 mM EDTA.
  • the protein solution was shock frozen and stored at -80DC.
  • KDR-GST-fusion protein expressed in insect cells (Sf-9) and purified by Glutathion affinity chromatography was used as kinase.
  • GST-KDR-fusion protein Proqinase, Freiburg i.Brsg., Germany
  • substrate for the kinase reaction the biotinylated copolymer poly-(Glu, Tyr; 4:1) which can be purchased e.g. from the company Cisbiointernational (Marcoule, France).
  • KDR enzyme amount depending on lot, adjusted to give an dF of about 300 - 400
  • 15 ⁇ l assay buffer [50 mM Hepes/NaOH pH7.0, 25 mM MgCI 2 , 5 mM MnCI 2 , 0.5 mM sodium ortho-vanadate, 1.0 mM dithiothreitol, 1 ⁇ M adenosine-tri-phosphate (ATP), 23.5 ⁇ g/ml substrate [biotinylated poly-(Glu, Tyr; 4:1)], 1.5% (v/v) dimethylsulfoxide].
  • the reaction was stopped by the addition of 5 ⁇ l of a solution of the detection reagents [0.3 ⁇ g/ml Eu-W1024-labeled antiphosphotyrosine antibody (PT66) (Perkin-Elmer) and 4.125 ⁇ g/ml SA-XL- 665 (Cisbiointernational, Marcoule, France)] in an aqueous EDTA -solution (250 mM EDTA, 0.1 % (w/v) bovine serum albumine in 100 mM HEPES/NaOH pH 7.0).
  • PT66 Eu-W1024-labeled antiphosphotyrosine antibody
  • SA-XL- 665 Cebiointernational, Marcoule, France
  • the resulting mixture was incubated further 2 h at 22°C to allow the binding of the biotinylated substrate and product to the SA-XL-665 and the EU labeled anti- phosphotyrosine antibody. Subsequently the amount of phosphate incorporated into the substrate was evaluated by resonance energy transfer measurement in a HTRF reader (Discovery, Perkin-Elmer).
  • the IC 50 values are determined from the inhibitor concentration that is necessary to inhibit the phosphate incorporation to 50% of the uninhibited incorporation after removal of the blank reading (EDTA-stopped reaction).
  • Figure 1 preferred compounds inhibiting preferentially Akt, Pdk kinases
  • Figure 3 preferred compounds inhibiting preferentially Akt and/or Pdk and VEGF-
  • R' is hydrogen or methyl
  • R' is Ci-eAlkyl
  • R' is Ci-eAlkyl and R" is cycloalkyl ring, heteroaryl or aryl; and R 1 , A and B are as described in the claims.
  • R" is Ci- ⁇ Alkyl and R and R 5 are as described in the claims.
  • Chloro[[(1 ,1-dimethylethoxy)carbonyl]amino]-sulfane dioxide was prepared by adding chlorosulfonyl isocyanate (32 mg, 0.23 mmol, 1.0 eq.) to a cooled solution of fetf-butyl alcohol (17 mg, 0.23 mmol, 1.0eq.) and dichloromethane (2 mL) in an ice-water bath. The resulting mixture was stirred at 0-5°C for 2-3hr.
  • Lithium Aluminum hydride (95%) (1.1 g, 27.5 mmol) was suspended in dry THF (20 mL) and cooled with an ice-water bath. A solution of 1 ,3-thiazol-4-acetonitrile (1.0 g, 8.06 mmol) in THF (10 mL) was added dropwise. The resulting mixture was stirred at room temperature overnight. To the reaction mixture was added water (1 mL), 15% NaOH (1 mL) followed by water (3 mL). The precipitate inorganic solid was filtered, then washed with ethyl acetate (100 mL).
  • the resulting solution was warmed to room temperature and stirred overnight.
  • the reaction mixture was concentrated under reduced pressure to remove dimethylformamide.
  • the crude residue was triturated in water to give a suspension.
  • the suspension was filtered and the filter cake was washed with water and air-dried (ca. 8 g).
  • Phenylmethyl[3-[[5-[[[[(1 ,1-dimethylethyl)dimethylsilyl]oxy]methyl]-2-[(3- nitrophenyl)amino]pyrimidin-4-yl]amino]propyl]carbamate (244 mg), dissolved in ethanol (30ml), was slowly added to a mixture of FeSO 4 heptahydrate (1.25 g), concentrated ammonia solution (25%; 1.25 ml) and water (5 ml). After refluxing for 3 h the mixture was filtered and the filter cake washed with ethyl acetate. The filtrate was washed with water and brine, dried (Na 2 SO ), filtered and evaporated to yield the crude title compound (230 mg), which was used in the next step without further purification.
  • N-(4- ⁇ 5-Bromo-4-[2-(3H-imidazol-4-yl)-ethylamino]-pyrimidin-2-ylamino ⁇ -phenyl)- trifluoro acetamide (1g, 1.9 mmole) was dissolved in THF (10 ml), MeOH (10 ml) and water (5 ml) and LiOH (455 mg) was added in one portion at room temperature. The reaction mixture was stirred at room temperature for two days, the solvent removed under reduced pressure. The residue was dissolved in ethyl acetate and water and extracted with ethyl acetate (3x). The combined organic layers were combined and dried over magnesium sulfate.
  • N-(4- ⁇ 5-Bromo-4-[2-(3H-imidazol-4-yl)-ethylamino]-pyrimidin-2-ylamino ⁇ -phenyl)- trifluoro acetamide (1g, 1.9 mmole) was dissolved in THF (10 ml), MeOH (10 ml) and water (5 ml) and LiOH (455 mg) was added in one portion at room temperature. The reaction mixture was stirred at room temperature for two days, the solvent removed under reduced pressure. The residue was dissolved in ethyl acetate and water and extracted with ethyl acetate (3x). The combined organic layers were combined and dried over magnesium sulfate.
  • N2-(4-Amino-phenyl)-5-bromo-N4-[2-(3H-imidazol-4-yl)-ethyl]-pyrimidine-2,4- diamine (1 g, 2.6 mmole, prepared according to procedure 21) was dissolved in MeOH (10 ml), butanal (0.261 ml, 2.9 mmole) was added at room temperature and the reaction mixture was stirred at room temperature for 20 minutes. Sodium cyanoborohydride (266 mg, 3.6 mmole) was added and the reaction mixture was stirred at room temperature overnight.
  • N2-(4-Amino-phenyl)-5- bromo-N4-[2-(3H-imidazol-4-yl)-ethyl]-pyrimidine-2,4-diamine 100 mg, 0.26 mmole, prepared according to procedure 21
  • the reaction was allowed to stir overnight.
  • extraction with bicarbonate solution and ethyl acetate (3x) the combined organic layers were dried over magnesium sulfate and the solvent was removed under reduced pressure.
  • N2-(4-Amino-phenyl)-5-bromo-N4-prop-2-ynyl-pyrimidine-2,4-diamine (100 mg, 0.3 mmole, prepared in analogy procedure 21) was dissolved in acetonitrile (10 ml) and allyl isothiocyanate (1 ml) was added at room temperature. The reaction mixture was heated under reflux for 3 hours, the solvent removed under reduced pressure and 1-Allyl-3-[4-(5-bromo-4-prop-2-ynylamino-pyrimidin-2- ylamino)-phenyl]-thiourea was crystallized from acetone / ethyl acetate / hexanes. Yield 37 mg. ESI-MS: 418.
  • N2-(4-Amino-phenyl)-5-bromo-N4-prop-2-ynyl-pyrimidine-2,4-diamine (100 mg, 0.3 mmole, prepared in analogy to procedure 21) was dissolved in acetonitrile (10 ml) and ethyl isocyanate (0.5 ml) was added at room temperature. The reaction mixture was heated under reflux for 5 hours and then cooled to room temperature and stirred overnight. The solid was filtered off and dried under high vaccum to provide 47 mg of 1-[4-(5-Bromo-4-prop-2-ynylamino-pyrimidin-2- ylamino)-phenyl]-3-ethyl-urea. ESI-MS: 390. A27
  • N2-(4-Amino-phenyl)-5-bromo-N4-prop-2-ynyl-pyrimidine-2,4-diamine (100 mg, 0.3 mmole, prepared in analogy to procedure 21) was dissolved in acetonitrile (10 ml) and triethylamine (1 ml) and 1 -Methyl-1 H-imidazole-4-sulfonyl chloride (120 mg, 0.66 mmole) was added at room temperature. The reaction mixture was stirred under reflux for 5 hours, the solvent was removed under reduced pressure and the crude product was purified by column chromatography on silica gel (ethyl acetate : hexanes 1 : 1).

Abstract

This invention relates to pyrimidine derivatives of general formula (I) as inhibitors of kinases, their production as well as their use as medications for treating various diseases.

Description

Chk-, Pdk- and Akt-lnhibitory Pyrimidines, Their Production and Use as
Pharmaceutical Agents
Description
This invention relates to pyrimidine derivatives, their production as well as their use as medications for treating various diseases.
The Chks (checkpoint kinases)-, Akts (protein kinases B) and Pdks (phosphoinositide-dependent kinases) are enzyme families that play an important role in the regulation of the cell cycle and thus is an especially advantageous target for the development of small inhibitory molecules. Akts and Pdks may be involved in common signal transduction pathways. Preferential inhibitors of the Chks and Akts and/or Pdks, particularly of Pdk1 can be used for treating cancer or other diseases that cause disruptions of cell proliferation.
Pyrimidines and analogs are already described as active ingredients, such as, for example, the 2-anilino-pyrimidines as fungicides (DE-A-4029650) or substituted pyrimidine derivatives for treating neurological or neurodegenerative diseases (WO 99/19305). As CDK-inhibitors, the most varied pyrimidine derivatives are described, for example bis(anilino)-pyrimidine derivatives (WO 00/12486), 2-amino-4-substituted pyrimidines (WO 01/14375), purines (WO 99/02162), 5-cyano-pyrimidines (WO 02/04429), anilinopyrimidines (WO 00/12486) and 2-hydroxy-3-N,N-dimethylaminopropoxy-pyrimidines (WO 00/39101).
Protein ligands and receptor tyrosine kinases that specifically regulate endothelial cell function are substantially involved in physiological as well as in disease-related angiogenesis. These ligand/receptor systems include the Vascular Endothelial Growth Factor (VEGF) and the Angiopoietin (Ang) families, and their receptors, the VEGF receptor family and the tyrosine kinase with immunoglobulin-like and epidermal growth factor homology domains (Tie) family. The members of the two families of receptor tyrosine kinases are expressed primarily on endothelial cells. The VEGF receptor family includes Flt1 (VEGF-R1), Flk1/KDR (VEGF-R2), and Flt4 (VEGF-R3). These receptors are recognized by members of the VEGF-related growth factors in that the ligands of Flt1 are VEGF and placenta growth factor (PIGF), whereas Flk1/KDR binds VEGF, VEGF-C and VEGF-D, and the ligands of Flt4 are VEGF-C and VEGF-D (Nicosia, Am. J. Pathol. 153, 11-16, 1998). The second family of endothelial cell specific receptor tyrosine kinases is represented by Tie1 and Tie2 (also kown as Tek). Whereas Tie1 remains an orphan receptor, three secreted glycoprotein ligands of Tie2, Ang1 , Ang2, and Ang3/Ang4 have been discovered (Davis et al., Cell 87, 1161-1169, 1996; Maisonpierre et al., Science 277, 55-60, 1997; Valenzuela et al, Proc. Natl. Acad. Sci. USA 96, 1904-1909, 1999; patents: US 5,521 ,073; US 5,650,490; US 5,814,464). Preferential inhibitors of the angiogenesis related kinases can be used for treating cancer or other diseases that are related to angiogenesis.
The object of this invention is to provide compounds that are inhibitors of cell cycle dependent kinases, in particular Chk, Akt, Pdk, CDK (cyclin dependent kinases) and/or angiogenesis related kinases, in particular VEGF-R (vascular endothelial growth factor receptor) kinases which have better properties than the inhibitors that are already known. The substances that are described here are more effective, since they already inhibit in the nanomolar range and can be distinguished from other already known Cdk-inhibitors such as, e.g., olomoucine and roscovitine.
It has now been found that the novel compounds of general formula I
Figure imgf000004_0001
A or B in each case independently of one another represent cyano, halogen, hydrogen, hydroxy, aryl or the group -NO2, -NH2, - NR3R4, -d-e-alkyl-NR^4, -N(C1-6-hydroxyalkyl)2l -NH-C(NH)-CH3, - NH(CO)-R5, -NHCOOR6, -NR7-(CO)-NR8R9, -NR7-(CS)-NR8R9, - COOR5 , -CO-NR8R9, -CONH-C1-6-alkyl-COOH, -SO2-CH3, 4- bromo-1 -methyl- 1 H-pyrazolo-3yl or represent Cι-6-alkyl optionally substituted in one or more places, the same way or differently with halogen, hydroxy, cyano or with the group -COOR5 , -CONR8R9, -NH2, -NH-SO2-CH3, - NR8R9, -NH-(CO)-R5, -NR7-(CO)-NR8R9, -SO2-NHR3, -O-(CO)-R5 or -O-(CO)-C1-6-alkyl-R5, X represents an oxygen atom or the group -NH- or -NR3R4,
R1 represents hydrogen, halogen, hydroxymethyl, C1-6-alkyl, cyano or the group -COOH, -COO-iso-propyl, -NO2, -NH-(CO)-(CH2)2- COOH or -NH-(CO)-(CH2)2-COO-Cι-6-alkyl, whereby the C1-6-alkyl can optionally be substituted in one or more places, in the same way or differently with halogen, R2 represents hydrogen or the group -NH-(CO)-aryl or d-β-alkyl optionally substituted in one or more places, the same way or differently with cyano, hydroxy, aryl, heteroaryl, C3-6- heterocycloalkylring, which can optionally be interrupted with one or more nitrogen atoms, or substituted with the group -NR8R9, - NH-(CO)-NR8R9, -NH-(CO)-S-Ci-6-alkyl, -NH-(CS)-NR8R9, -NH- (CO)O-CH2-phenyl, -NH-(CO)H, -NH(CO)-R5, -NH(CO)-OR5, - (CO)-NH-NH2, -(CO)-NH-CH2-(CO)-NH2, -(CO)-NH-Cι-6-alkyl, -
COOH,
Figure imgf000006_0001
whereby the aryl or the heteroaryl can optionally be substituted in one or more places, the same or differently with halogen, hydroxy, d-e-alkyl, -NH2, -NH-(CO)-CH2-NH2, -NO2, -(CO)-C(CH2)-C2H5, - COOR6, -COOC(CH3)3, or represents C3-alkinyl, R3 and R4 in each case independently of one another represent hydrogen or Cι-6-alkyl optionally substituted in one or more places, the same way or differently with hydroxy, phenyl or hydroxyphenyl, or R3 or R4 together form a C3-6-heterocycloalkylring containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C3-6- heterocycloalkylnng can optionally be substituted with Cι-6-alkyl, d-e-alkyl-COOH or Cι-6-alkyl-NH2, R5 represents hydrogen, d-6-alkyl, d-6-alkoxy, C2-6-alkenyl, C3-6- cycloalkylring, aryl, heteroaryl, the group -(CO)-NH2 or C3-6- heterocycloalkylring that can optionally be interrupted with one or more nitrogen and/or oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring and d-6-alkyl, C2-6-alkenyl, C3-6-cycloalkylring, C3-6- heterocycloalkylnng defined above, aryl or heteroaryl can optionally be substituted in one or more places, the same way or differently with halogen, hydroxy, d-β-alkyl, Cι-6-alkoxy, C3-6- cycloalkyl, C3-6-heterocycloalkylring defined above, aryl, heteroaryl or with the group -NR8R9, -NO2, -NR7-(CO)-R5, -NH(CO)-C1-6- alkyl-NH-(CO)-d-6-alkyl, -NR7-(CO)-NR8R9, -CO-CH3, -COOH, -
CO-NR8R9, -SO2-aryl, -SH, -S-Ci-e-alkyl, -SO2-NR8R9, whereby aryl itself can optionally be substituted in one or more places, the same way or differently with halogen, hydroxy, Chalky! or Ci-6-alkoxy, R6 represents Cι-6-alkyl, C2-6-alkenyl or phenyl, whereby d-β-alkyl may optionally be substituted with C3-6- heterocycloalkylring that can optionally be interrupted with one or more nitrogen and/or oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, R7 represents hydrogen or d-β-alkyl, R8or R9 in each case independently of one another represent hydrogen, d-6-alkyl, C2-6-alkenyl, C3-6-cycloalkyl, aryl or heteroaryl or the group
Figure imgf000008_0001
whereby Cι-6-alkyl, C2-6-alkenyl, C3-6-cycloalkyl, aryl or heteroaryl can optionally be substituted in one or more places, the same way or differently with halogen, heteroaryl, hydroxy, d-6-alkoxy, hydroxy-Ci-e-alkoxy or the group -COOH, -NO2, -NR8R9, -N(d-6- alkyl)2 or with a C3-6-heterocycloalkylring can optionally be interrupted with one or more nitrogen and/or oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, or R8 and R9 together form a C3-6-heterocycloalkylring containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C3-β- heterocycloalkylring can optionally be substituted in one or more places, the same way or differently with hydroxy or the group - NR8R9, -NH(CO)-R5, hydroxy-C1-6-alkyl or -COOH and
R10 represents -SO2-aryl, -SO2-heteroaryl or -SO2-NH2 or -SO2-Cι-6- alkyl, whereby the aryl can be substituted with -d-6-alkyl, with the following provisos: whereby if X represents -NR3R4 then R2 does not represent a substituent, whereby if A and B represent hydrogen, X represents -NH- and R2 represents d-e-alkyl, then R1 represents -NH-(CO)-CH(NH2)-(CH2)2-COOH or -NH-
(CO)-CH(NH2)-(CH2)2-COOC2H5, whereby if A represents-(CO)-OC2H5 or hydroxy, B represents hydrogen, X represents oxygen, R1 represents halogen, then R2 represents C3-alkinyl, whereby if A represents -(CO)-OC2H5 or hydroxy, B represents hydrogen, X represents -NH-, R1 represents -NO2, then R2 represents C3-alkinyl, whereby if A represents -(CO)-OCH3, then X represents oxygen, R1 represents halogen, R2 represents
C3-alkinyl and B represenst -NH2, -NHC2H4OH, -N(C2H4OH)2, -
NH-(CO)-CH2-O(CO)CH3, whereby if A represents -(CO)-OCH3, then X represents -NH-, R1 represents halogen, R2 represents -
C2H -imidazolyl and B represenst hydrogen -NH2, whereby if A represents -NHS02-CH3, then B represents hydrogen, X represents -NH-, R1 represents halogen and R2 represents -C2H4-imidazolyl, whereby if R1 represents -COO-iso-propyl, then X represents -NH- and R2 represents C3-alkinyl and A or B independently of one another represent the group -NO2 or -NH-
(CO)-CF3, whereby if R1 represents halogen, X represents -NH-, B represents hydrogen and R2 represents Cι-6-alkyl substituted with -NH2, then A represents -NH-(CO)-C6-cycloalkyl-NH2, whereby if R1 represents halogen, X represents -NH-, B represents -S-CH3 and R2 represents imidazolyl, then A represents the group
Figure imgf000009_0001
as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof are capable of inhibiting kinases which are involved in the regulation of the cell cycle, particulary Chks, Akt, Pdks and/or Cdks as well as angiogenesis related kinases, particulary VEGF-R kinases.
A more detailed explanation of the terms used in the claims and the description is given in the following:
As used herein the singular forms "a", "and", and "the" include plural referents unless the context clearly dictates otherwise. For example, "a compound" refers to one or more of such compounds, while "the enzyme" includes a particular enzyme as well as other family members and equivalents thereof as known to those skilled in the art.
Preferred aspects of the present invention are described in the claims. A more detailed explanation of the terms used in the claims is given in the following:
"Alkyl" is defined in each case as a straight-chain or branched alkyl radical, such as, for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, hexyl, heptyl, octyl, nonyl and decyl.
"Alkoxy" is defined in each case as a straight-chain or branched alkoxy radical, such as, for example, methyloxy, ethyloxy, propyloxy, isopropyloxy, butyloxy, isobutyloxy, sec-butyloxy, tert-butyloxy, pentyloxy, isopentyloxy, or hexyloxy.
"Hydroxy-Alkoxy" is defined in each case as a straight-chain or branched alkoxy radical, such as, for example, methyloxy, ethyloxy, propyloxy, isopropyloxy, butyloxy, isobutyloxy, sec-butyloxy, tert-butyloxy, pentyloxy, isopentyloxy, or hexyloxy is substituted one or more times with hydroxy.
"Alkylthio" is defined in each case as a straight-chain or branched alkylthio radical, such as, for example, methylthio, ethylthio, propylthio, isopropylthio, butylthio, isobutylthio, sec-butylthio, tert-butylthio, pentylthio, isopentylthio or hexylthio. "Cycloalkyl" is defined in general as monocyclic alkyl rings, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl or cyclodecyl, but also bicyclic rings or tricyclic rings such as, for example, norbornyl, adamantanyl, etc.
The ring systems, in which optionally one or more possible double bonds can be contained in the ring, are defined as, for example, cycloalkenyls, such as cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, or cycloheptenyl, whereby the linkage can be carried out both to the double bond and to the single bonds.
If R3 and R4 or R8 and R9 as defined in the claims, in each case independently of one another, together form a C3-Cι0-cycloalkyl ring, which optionally can be interrupted by one or more heteroatoms, such as nitrogen atoms, oxygen atoms and/or sulfur atoms, and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally one or more possible double bonds can be contained in the ring, however, the above-mentioned definitions are also intended to include heteroaryl radical or heterocycloalkyl and heterocycloalkenyl. In terms of this invention interrupted can mean either that the heteroatoms in addition to the carbon atoms form the ring or that the heteroatoms are substitutes for one or more carbon atoms.
"Halogen" is defined in each case as fluorine, chlorine, bromine or iodine.
The "alkenyl" substituents in each case are straight-chain or branched, whereby, for example, the following radicals are meant: vinyl, propen-1-yl, propen-2-yl, but-1-en-1-yl, but-1-en-2-yl, but-2-en-1-yl, but-2-en-2-yl, 2-methyl-prop-2-en-1-yl, 2-methyl-prop-1-en-1-yl, but-1-en-3-yl, ethinyl, prop-1-in-1-yl, but-1-in-1-yl, but-2-in-1-yl, but-3-en-1-yl, and allyl.
"Alkinyl" is defined in each case as a straight-chain or branched alkinyl radical that contains 2-6, preferably 2-4 C atoms. For example, the following radicals can be mentioned: acetylene, propin-1-yl, propin-3-yl, but-1-in-1-yl, but-1-in-4-yl, but-2-in-1-yl, but-1-in-3-yl, etc.
The "aryl" radical in each case comprises 3-16 carbon atoms and in each case can be benzocondensed.
For example, there can be mentioned: cyclopropenyl, cyclopentadienyl, phenyl, tropyl, cyclooctadienyl, indenyl, naphthyl, azulenyl, biphenyl, fluorenyl, anthracenyl, etc.
The "heteroaryl" radical in each case comprises 3-16 ring atoms, and instead of the carbon can contain one or more heteroatoms that are the same or different, such as oxygen, nitrogen or sulfur, in the ring, and can be monocyclic, bicyclic, or tricyclic and in addition in each case can be benzocondensed.
For example, there can be mentioned:
Thienyl, furanyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, etc. and benzo derivatives thereof, such as, e.g., benzofuranyl, benzothienyl, benzoxazolyl, benzimidazolyl, indazolyl, indolyl, isoindolyl, etc.; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, etc. and benzo derivatives thereof, such as, e.g., quinolyl, isoquinolyl, etc., or azocinyl, indolizinyl, purinyl, etc. and benzo derivatives thereof; or quinolinyl, isoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, naphthyridinyl, pteridinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, xanthenyl, oxepinyl, etc.
"Heterocycloalkyl" stands for an alkyl ring that comprises 3- 6 carbon atoms, which can optionally be interrupted with one or more nitrogen and/or oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring. In terms of this invention interrupted can mean either that the heteroatoms in addition to the carbon atoms form the ring or that the heteroatoms are substitutes for one or more carbon atoms.
For purposes of this invention, the heterocycloalkyl radical may be a monocyclic, or bicyclic ring system, which may include fused or bridged ring systems; and additionally the nitrogen or sulfur atoms in the heterocyclyl radical may be optionally oxidized; the nitrogen atom may be optionally quatemized; and the heterocyclyl radical may be aromatic or partially or fully saturated.
As heterocycloalkyls, there can be mentioned, e.g.: oxiranyl, oxethanyl, aziridinyl, azetidinyl, tetrahydrofuranyl, pyrrolidinyl, pyrrolidinonyl, dioxolanyl, imidazolidinyl, imidazolidinonyl, thiazolidiononyl, pyrazolidinyl, pyrazolidinonyl, dioxanyl, piperidinyl, piperidinonyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, trithianyl, quinuclidinyl, oxazolidinyl, oxazolidinonyl, hydantoin, pyran, thiin, dihydroacet, etc.
As used herein, "suitable conditions" for carrying out a synthetic step are explicitly provided herein or may be discerned by reference to publications directed to methods used in synthetic organic chemistry. The reference books and treatise set forth above that detail the synthesis of reactants useful in the preparation of compounds of the present invention, will also provide suitable conditions for carrying out a synthetic step according to the present invention. As used herein, "methods known to one of ordinary skill in the art" may be identified though various reference books and databases. Suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds of the present invention, or provide references to articles that describe the preparation, include for example, "Synthetic Organic Chemistry", John Wiley & Sons, Inc., New York; S. R. Sandier et al., "Organic Functional Group Preparations," 2nd Ed., Academic Press, New York, 1983; H. O. House, "Modern Synthetic Reactions", 2nd Ed., W. A. Benjamin, Inc. Menlo Park, Calif. 1972; T. L. Gilchrist, "Heterocyclic Chemistry", 2nd Ed., John Wiley & Sons, New York, 1992; J. March, "Advanced Organic Chemistry: Reactions, Mechanisms and Structure", 4th Ed., Wiley-lnterscience, New York, 1992. Specific and analogous reactants may also be identified through the indices of known chemicals prepared by the Chemical Abstract Service of the American Chemical Society, which are available in most public and university libraries, as well as through on-line databases (the American Chemical Society, Washington, D.C. may be contacted for more details). Chemicals that are known but not commercially available in catalogs may be prepared by custom chemical synthesis houses, where many of the standard chemical supply houses (e.g., those listed above) provide custom synthesis services.
"Stable compound" and "stable structure" are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
"Mammal" includes humans and domestic animals, such as cats, dogs, swine, cattle, sheep, goats, horses, rabbits, and the like.
"Optional" or "optionally" means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, "optionally substituted aryl" means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
"Pharmaceutically acceptable carrier, diluent or excipient" includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
"Pharmaceutically acceptable salt" includes both acid and base addition salts. "Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like.
"Pharmaceutically acceptable base addition salt" refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2- dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like. Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline caffeine, N-methyl-glucamine, dimethyl-glucamine, ethyl-glucamine, lysine,
1 ,6-hexadiamine, ethanol-amine, glucosamine, sarcosine, serinol, tris-hydroxy-methyl-amino-methane, aminopropane diol, Sovak base, and 1-amino-2,3,4-butanetriol.
As used herein, compounds which are "commercially available" may be obtained from standard commercial sources including Acros Organics (Pittsburgh PA), Aldrich Chemical (Milwaukee Wl, including Sigma Chemical and Fluka), Apin Chemicals Ltd. (Milton Park UK), Avocado Research (Lancashire U.K.), BDH Inc. (Toronto, Canada), Bionet (Cornwall, U.K.), Chemservice Inc. (West Chester PA), Crescent Chemical Co. (Hauppauge NY), Eastman Organic Chemicals, Eastman Kodak Company (Rochester NY), Fisher Scientific Co. (Pittsburgh PA), Fisons Chemicals (Leicestershire UK), Frontier Scientific (Logan UT), ICN Biomedicals, Inc. (Costa Mesa CA), Key Organics (Cornwall U.K.), Lancaster Synthesis (Windham NH), Maybridge Chemical Co. Ltd. (Cornwall U.K.), Parish Chemical Co. (Orem UT), Pfaltz & Bauer, Inc. (Waterbury CN), Polyorganix (Houston TX), Pierce Chemical Co. (Rockford IL), Riedel de Haen AG (Hannover, Germany), Spectrum Quality Product, Inc. (New Brunswick, NJ), TCI America (Portland OR), Trans World Chemicals, Inc. (Rockville MD), and Wako Chemicals USA, Inc. (Richmond VA). As used herein, "suitable conditions" for carrying out a synthetic step are explicitly provided herein or may be discerned by reference to publications directed to methods used in synthetic organic chemistry. The reference books and treatise set forth above that detail the synthesis of reactants useful in the preparation of compounds of the present invention, will also provide suitable conditions for carrying out a synthetic step according to the present invention. As used herein, "methods known to one of ordinary skill in the art" may be identified though various reference books and databases. Suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds of the present invention, or provide references to articles that describe the preparation, include for example, "Synthetic Organic Chemistry", John Wiley & Sons, Inc., New York; S. R. Sandier et al., "Organic Functional Group Preparations," 2nd Ed., Academic Press, New York, 1983; H. O. House, "Modern Synthetic Reactions", 2nd Ed., W. A. Benjamin, Inc. Menlo Park, Calif. 1972; T. L. Gilchrist, "Heterocyclic Chemistry", 2nd Ed., John Wiley & Sons, New York, 1992; J. March, "Advanced Organic Chemistry: Reactions, Mechanisms and Structure", 4th Ed., Wiley-lnterscience, New York, 1992. Specific and analogous reactants may also be identified through the indices of known chemicals prepared by the Chemical Abstract Service of the American Chemical Society, which are available in most public and university libraries, as well as through on-line databases (the American Chemical Society, Washington, D.C. may be contacted for more details). Chemicals that are known but not commercially available in catalogs may be prepared by custom chemical synthesis houses, where many of the standard chemical supply houses (e.g., those listed above) provide custom synthesis services.
"Prodrugs" is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of the invention. Thus, the term "prodrug" refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable. A prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound of the invention. Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam). A discussion of prodrugs is provided in Higuchi, T., et al., "Pro-drugs as Novel Delivery Systems," A.C.S. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein.
The term "prodrug" is also meant to include any covalently bonded carriers which release the active compound of the invention in vivo when such prodrug is administered to a mammalian subject. Prodrugs of a compound of the invention may be prepared by modifying functional groups present in the compound of the invention in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound of the invention. Prodrugs include compounds of the invention wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups in the compounds of the invention and the like.
"Therapeutically effective amount" refers to that amount of a compound of formula (I) which, when administered to a mammal, preferably a human, is sufficient to effect treatment, as defined below, for a disease-state alleviated by the inhibition of AKT-, PDK-, CHK-, CDK- or VEGF-R- acitivity, such as cancer.
The amount of a compound of formula (I) which constitutes a "therapeutically effective amount" will vary depending on the compound, the condition and its severity, and the age of the mammal to be treated, but can be determined routinely by one of ordinary skill in the art having regard to his own knowledge and to this disclosure.
"Treating" or "treatment" as used herein covers the treatment of disease-states alleviated by the inhibition of AKT-, PDK-, CHK-, CDK- or VEGF-R- activity, such as cancer, as disclosed herein, in a mammal, preferably a human, and includes:
(i) preventing the disease-state from occurring in a mammal, in particular, when such mammal is predisposed to the disease-state but has not yet been diagnosed as having it;
(ii) inhibiting the disease-state, i.e.., arresting its development; or (iii) relieving the disease-state, i.e.., causing regression of the condition. The compounds of formula (I), or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or ( )- for amino acids. The present invention is meant to include all such possible isomers, as well as, their racemic and optically pure forms. Optically active (+) and (-), (f?)- and (S)-, or (D)- and ( )- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, such as reverse phase HPLC. When the formulae described herein contain olefinic double bonds or other centers of geometric asymmetry, unless specified otherwise, it is intended that the formulae include both E and Z geometric isomers, as well as all tautomeric forms. In addition, all compound names herein, unless specified otherwise, are intended to include all single enantiomers, diastereomers, and mixtures thereof, as well as racemic and non-racemic mixtures.
Compounds which preferentially inhibit AKT and/or PDK kinases are the compounds of formula I in which
A or B in each case independently of one another represent cyano, halogen, hydrogen, hydroxy, tetrazolyl or the group -NH2, -NR3R4,
-d-6-alkyl-NR3R4, -NH-C(NH)-CH3, -NH(CO)-R5, -NHCOOR6, - NR7-(CO)-NR8R9, - d-e-alkyl-COOH, -COOH , -CONH2, -CONH- Ci-e-alkyl-COOH, or represent d-6-alkyl optionally substituted in one or more places, the same way or differently with halogen, hydroxy or with the group -COOH , -CONR8R9, -NH-SO2-CH3 or -NR8R9, X represents the group -NH- or -NR3R4,
R1 represents cyano, hydrogen, halogen or Cι-6-alkyl, whereby the Ci.
6-alkyl can optionally be substituted in one or more places, in the same way or differently with halogen,
R2 represents hydrogen or the group -NH-(CO)-aryl or -C1-6-alkyl optionally substituted in one or more places, the same way or differently with cyano, hydroxy, aryl , heteroaryl, C3-6- heterocycloalkylring which can be optionally be interrupted in one or more places with one or more nitrogen atoms, or substituted with the group -NR8R9, -NH-(CO)-NR8R9, -NH-(CO)-S-C1-6-alkyl, - NH-(CS)-NR8R9, -NH(CO)-R5, -NH(CO)-OR5, -(CO)-NH-NH2, - (CO)-NH-CH2-(CO)-NH2, -(CO)-NH-C1-6-alkyl, -COOH whereby the aryl or the heteroaryl can optionally be substituted in one or more places, the same way or differently with hydroxy, Cι-6-alkyl, -NH2, - NH-(CO)-CH2-NH2, -NO2, -COOR6,
Figure imgf000020_0001
R3 or R4 in each case independently of one another represent hydrogen, Cι-6-alkyl optionally substituted in one or more places, the same way or differently with hydroxy, phenyl or hydroxyphenyl, or
R3 and R4 together form a C3-6-heterocycloalkylring containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C3-6- heterocycloalkylring can optionally be substituted with d-6-alkyl, C1-6-alkyl-COOH or C1-6-alkyl-NH2, R5 represents hydrogen, d-6-alkyl, Cι-6-alkoxy, C2-6-alkenyl, C3-6- cycloalkylring, heteroaryl , the group -(CO)-NH2 or C3-6- heterocycloalkylring that can optionally be interrupted with one or more nitrogen and/or oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring and Cι-6-alkyl, C2-6-alkenyl, C3-6-heterocycloalkylring define above, aryl or heteroaryl can optionally be substituted in one or more places, the same way or differently with halogen, hydroxy, d-6- alkyl, Cι-6-alkoxy, C3-6-cycloalkyl, C3-6-heterocycloalkylring define above, aryl, heteroaryl or with the -NR8R9, -NO2, -NR7-(CO)-R5, - NH(CO)-Cι-6-alkyl-NH-(CO)-Cι-6-alkyl, -NR7-(CO)-NR8R9, -CO-CH3,
-COOH, -CO-NR8R9, -SO2-aryl, -SH, -S-d-6-alkyl, -SO2-NR8R9, whereby aryl itself can optionally be substituted in one or more places, the same way or differently with halogen or hydroxy, Chalky! or Cι-6-alkoxy, R7 represents hydrogen or Ci-β-alkyI,
R8or R9 in each case independently of one another represent hydrogen,
Cι-6-alkyl, aryl or heteroaryl or the group R10, whereby d-6-alkyl, aryl or heteroaryl can optionally be substituted in one or more places, the same way or differently with halogen, heteroaryl, hydroxy, C1-6-alkoxy, hydroxy-d-6-alkoxy or with the group -
COOH, -NO2, or a C3-6-heterocycloalkylring can optionally be interrupted with one or more nitrogen and/or oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring or R8 and R9 together form a C3-6-heterocycloalkylring containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C3-6- heterocycloalkylring can optionally be substituted in one or more places, the same way or differently with hydroxy, hydroxy-C1-6-alkyl or the group -NR8R9, -NH(CO)-R5 or -COOH and
R10 represents -SO2-NH2, -SO2-C1-6-alkyl, -SO2-aryl, or -SO2- heteroaryl, whereby the aryl can be substituted with -d-6-alkyl, as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
Even more preferred are the compounds of formula I, which inhibit preferentially AKT and/or PDK kinases in which
A or B in each case independently of one another represent hydrogen, tetrazolyl or the group -N(CH3)2, -NH-(CO)-pyrrolidinyl, -NH-(CO)- pentyl, -NH-(CO)-hexyl, -NH-(CO)-hexyl-NH2, -NH-(CO)-C3H7, - NH-(CO)-CH2-phenyl, -NH-(CO)-CH2-NH2, -NH-(CO)-C2H4-NH2, - NH-(CO)-CH(NH2)-CH3, -NH-(CO)-CH(NH2)-hydroxyphenyl, -NH-
(CO)-CH(NH2)-CH2-phenyl, -NH-(CO)-CH(NH2)-CH2- hydroxyphenyl, -NH-(CO)-CH(NH-(CO)-CH3)-CH2-phenyl, -NH- (CO)-CH2-NH-(CO)-CH3, -NH-(CO)-N(C2H5)(C2H4-piperidinyl), -NH- (CO)-N(CH3)(C2H4-piperidinyl), -NH-(CO)-CH2-NH(CH3), -CH2- N(CH3)2, -NH-(CO)NH-CH2-COOH , hydantoinyl, -CH2-COOH whereby the pyrrolidinyl can optionally be substituted with hydroxy or the group -NH2, -N(CH3)2 or -NH-(CO)-CH3, and whereby hydantoinyl can be substituted with -CH3, -CH2- COOH, or -(CO)-thiazolidinonyl, X represents or the group -NH-,
R1 represents halogen and
R2 represents hydrogen or the group -NH-(CO)-phenyl or -C2H -, -C3H6- both can optionally be substituted in one or more places, the same way or differently with cyano, hydroxy, phenyl, naphthyl, imidazolyl, thiazolyl, pyridyl, 2-oxazolinyl, piperidinyl, - NH2, -NH-CH2-thienyl, -NH-pyridinyl-NO2, -NH-thiazolyl, -SO2- thienyl, -SO2-NH2, -SO2-CH3, -SO2-C3H7, pyrrolidinonyl substituted with -COOH, -NH-(CO)-NH-thienyl, -NH-(CO)-NH-phenyl, -NH- (CO)-NH- C2H5, -NH-(CO)-C(CH3)3, -NH-(CO)-S-C2H5, -NH-(CS)- NH- C2H5, -NH-(CO)-C2H5, -NH-(CO)-thienyl, -(CO)-NH-NH2, - (CO)-NH-CH2-(CO)-NH2, -(CO)-NH-C2H5, -COOH whereby the phenyl or the imidazolyl, thiazolyl can optionally be substituted in one or more places, the same way or differently with hydroxy, - CH3, -NH-(CO)-CH2-NH2, -COOC2H5, -COOC(CH3)3,
Figure imgf000023_0001
as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
Even more preferred are compounds of general formula (I), which inhibit preferentially AKT and/or PDK kinases in which
A or B in each case independently of one another represent hydrogen or the group -NH-(CO)-pyrrolidinyl, -NH-(CO)-piperidinyl, -NH-(CO)- morpholinyl, -NH-(CO)-hexyl-NH2, -NH-(CO)-CH(NH2)- hydroxyphenyl, -NH-(CO)-CH(NH2)-CH2-hydroxyphenyl, hydantoin optionally substituted with -CH3, X represents or the group -NH-,
R1 represents halogen and
R2 represents hydrogen, -C2H4-imidazolyl or -C3H wich can optionally be substituted in one or more places, the same way or differently with the group -NH-CH2-thienyl, -NH-(CO)-C2H5, -NH-(CO)- C(CH3)3,
as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
In particular the following compounds of general formula (I) are preferred to inhibit preferentially AKT and/or PDK kinases:
N-[3-[[5-bromo-4-[[3-[[[1-
(trifluoromethyl)cyclobutyl]carbonyl]amino]propyl]amino]-2- pyrimidinyl]amino]phenyl]-1-pyrrolidinecarboxamide,
N-[3-[[5-bromo-4-[[3-[[1-oxo-3-(phenylsulfonyl)propyl]amino]propyl]amino]-2- pyrimidinyl]amino]phenyl]-1-pyrrolidinecarboxamide,
N-[3-[[5-bromo-2-[[3-[(1-pyrrolidinylcarbonyl)amino]phenyl]amino]-4- pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide,
N-[3-[[4-[[3-[[(1-aminocyclopentyl)carbonyl]amino]propyl]amino]-5-bromo-2- pyrimidinyl]amino]phenyl]-1-pyrrolidinecarboxamide,
N-[3-[[4-[[3-[[(1-aminocyclobutyl)carbonyl]amino]propyl]amino]-5-iodo-2- pyrimidinyl]amino]phenyl]-1 -pyrrolidinecarboxamide,
N1-[3-[[5-bromo-2-[[3-[(1-pyrrolidinylcarbonyl)amino]phenyl]amino]-4- pyrimidinyl]amino]propyl]-1 , 1 -cyclopentanedicarboxamide,
(4R)-Λ/-[3-[[5-bromo-2-[[3-(2,5-dioxo-1-imidazolidinyl)phenyl]amino]-4- pyrimidinyl]amino]propyl]-2-oxo-4-thiazolidinecarboxamide, (4R)-N-[3-[[5-bromo-2-[[3-(3-methyl-2,5-dioxo-1-imidazolidinyl)phenyl]amino]-4- pyrimidinyl]amino]propyl]-2-oxo-4-thiazolidinecarboxamide,
3-[3-[[5-bromo-4-[[2-(1 H-imidazol-4-yl)ethyl]amino]-2-pyrimidinyl]amino]phenyl]-
2,4-imidazolidinedione,
3-[3-[[5-bromo-4-[[2-(1 H-imidazol-4-yl)ethyl]amino]-2-pyrimidinyl]amino]phenyl]- 1-methyl-2,4-imidazolidinedione,
N'-[3-[[5-bromo-4-[[2-(1H-imidazol-4-yl)ethyl]amino]-2-pyrimidinyl]amino]phenyl]-
N-ethyl-N-[2-(1-piperidinyl)ethyl]-urea,
N-[3-[[5-bromo-4-[[3-[(2,2-dimethyl-1-oxopropyl)amino]propyl]amino]-2- pyrimidinyl]amino]phenyl]-1 -pyrrolidinecarboxamide, N-[3-[[2-[[3-[[(2S)-2-amino-3-(4-hydroxyphenyl)-1 - oxopropyl]amino]phenyl]amino]-5-bromo-4-pyrimidinyl]amino]propyl]-2,2- dimethyl-propanediamide,
N-[3-[[2-[[3-[[(1-aminocyclohexyl)carbonyl]amino]phenyl]amino]-5-bromo-4- pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide, N-[3-[[2-[[3-[[(2S)-2-amino-2-phenylacetyl]amino]phenyl]amino]-5-bromo-4- pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide,
N-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5-bromo-
4-pyrimidinyl]amino]propyl]-5-oxo-2-pyrrolidinecarboxamide,
N-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5-bromo- 4-pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide,
N1-[3-[[5-bromo-2-[[3-[[(2S)-2-pyrrolidinylcarbonyl]amino]phenyl]amino]-4- pyrimidinyl]amino]propyl]- 1 ,1-cyclopropanedicarboxamide,
N-[3-[[5-bromo-2-[[3-(2,5-dioxo-1-imidazolidinyl)phenyl]amino]-4- pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide,
/V-(3-((5-bromo-4-((2-(7H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-4-morpholinecarboxamide,
Λ/-(3-((5-bromo-4-((2-( H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-1 -pyrrolidinecarboxamide,
Λ/-(3-((5-bromo-4-((3-((2-thienylcarbonyl)amino)propyl)amino)-2- pyrimidinyl)amino)phenyl)-1-pyrrolidinecarboxamide,
/V1-(3-((5-bromo-2-((3-((1-pyrrolidinylcarbonyl)amino)phenyl)amino)-4- pyrimidinyl)amino)propyl)-1 ,1-cyclopropanedicarboxamide, Λ/-(3-((5-bromo-4-((3-((1-oxopropyl)amino)propyl)amino)-2- pyrimidinyl)amino)phenyl)-1-pyrrolidinecarboxamide,
Λ/-(3-((5-iodo-4-((3-((2-thienylcarbonyl)amino)propyl)amino)-2-pyrimidinyl)- amino)phenyl)-1 -pyrrolidinecarboxamide,
N-[3-[[5-bromo-4-[[3-[[[(2S)-5-oxo-2-pyrrolidinyl]carbonyl]amino]propyl]amino]-2- pyrimidinyl]amino]phenyl]-1 -pyrrolidinecarboxamide,
N-[3-[[5-bromo-4-[[3-[[[(2S)-4-oxo-2-azetidinyl]carbonyl]amino]propyl]amino]-2- pyrimidinyl]amino]phenyl]-1-pyrrolidinecarboxamide,
(4R)-N-[3-[[5-bromo-2-[[3-[(1-pyrrolidinylcarbonyl)amino]phenyl]amino]-4- pyrimidinyl]amino]propyl]-2-oxo-4-thiazolidinecarboxamide or N-[3-[[4-[[3-[[(1-aminocyclobutyl)carbonyl]amino]propyl]amino]-5-bromo-2- pyrimidinyl]amino]phenyl]-1-pyrrolidinecarboxamide.
Preffered are also compounds of general formula (I), which inhibit preferentially
Chk kinases in which
A or B in each case independently of one another represent hydrogen or the group -NO2, -NH2, -NR3R4, -N(C1-6-hydroxyalkyl)2, -NH(CO)- R5, -NHCOOR6, -NR7-(CO)-NR8R9, -NR7-(CS)-NR8R9, -COOR5, - CO-NR8R9, -SO2-CH3, 4-bromo-1-methyl-1H-pyrazolo-3yl or d-e-alkyl optionally substituted in one or more places, the same way or differently with cyano, halogen, hydroxy or the group -NH2, -NH-(CO)-R5, -SO2-NHR3, -COOR5, -CONR8R9, -O-(CO)-R5, -O- (CO)-Cι-6-alkyl-R5, X represents an oxygen atom or the group -NH-,
R1 represents hydrogen, halogen, hydroxymethyl or the group -
COOH, -COO-iso-propyl, -NO2, -NH-(CO)-(CH2)2-COOH or -NH- (CO)-(CH2)2-COO-C1-6-alkyl, R2 represents Cι-6-alkyl optionally substituted in one or more places, the same way or differently with hydroxy, imidazolyl or the group - NH2, -NH-(CO)O-CH2-phenyl, -NH-(CO)H, -NH-(CO)-phenyl, -NH- (CO)-CH2-O-phenyl, -NH-(CO)-CH2-phenyl, -NH-(CO)- CH(NH2)CH2-phenyl, -NH-(CO)-CH2-CH(CH3)-phenyl, -NH-(CO)- CH(NH2)-(CH2)2-COOH,
Figure imgf000028_0001
, whereby the phenyl can optionally be substituted in one or more places, the same or differently with halogen, d-6-alkyl or -(CO)- C(CH2)-C2H5, or represents C3-alkinyl,
R3 or R4 in each case independently of one another represent hydrogen or d-β-alkyl optionally substituted in one or more places, the same way or differently with hydroxy, phenyl or hydroxyphenyl, or R3 and R4 together form a C3-6-heterocycloalkylring containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupoted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C3-6- heterocycloalkylring can optionally be substituted with C1-6-alkyl, d-6-alkyl-COOH or Cι-6-alkyl-NH2, R5 represents Cι-6-alkyl, C2-6-alkenyl, C3-6-cycloalkyl or phenyl each can optionally be substituted in one or more places, the same way or differently with halogen, hydroxy, phenyl or with the group -NH2, -NH(CO)-O-Cι-6-alkyl, whereby phenyl itself can optionally be substituted in one or more places, the same way or differently with halogen, hydroxy or Cι-6-alkyl, R6 represents d-β-alkyl, C2-6-alkenyl or phenyl,
R7 represents hydrogen or d-β-alkyl and
R8or R9 in each case independently of one another represent hydrogen, d-6-alkyl, C2.6-alkenyl, C3-6-cycloalkyl, aryl or phenyl, whereby aryl or phenyl can optionally be substituted in one or more places, the same way or differently with hydroxy or the group -NO2 or -N(Cι-6- alkyl)2 or R8 and R9 together form a C3.6-heterocycloalkylring containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C3-6- heterocycloalkylring can optionally be substituted with the group - NH2, as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
Even more preferred are those compounds of general formula (I), which inhibit preferentially Chk kinases in which
A or B in each case independently of one another represent hydrogen or the group -NH-C2H4-OH, -NH-CH2-hydroxyphenyl, -NH-(CO)- pyrrolidinyl, -NH-(CO)-CH(NH2)-CH2-phenyl, -NH-(CO)-pentyl-NH2, -NH-(CO)-hexyl-NH2, -NH-(CO)-CH2-NH2, -NH-(CO)-CH(NH2)- hydroxyphenyl, -NH-(CO)-CH2-hydroxyphenyl, -NH-(CO)-CH2- methylphenyl, -NH-(CO)-C2H4-dihydroxyphenyl, -NH-(CO)- CH(OH)-phenyl, -NH-(CO)-CH(NH2)-CH2(OH), -NH-(CO)- C(CH3)2NH2, -NH-(CO)-NH(C2H5), -CH2OH, -(CO)-NH-cyclopropyl,
-(CO)-NH-CH(CH3)2, whereby the pyrrolidinyl can optionally be substituted with hydroxy or the group -NH2, X represents an oxygen atom or the group -NH-, R1 represents halogen or hydroxymethyl and
R2 represents -C2H5 optionally substituted in one or more places, the same way or differently with hydroxy, imidazolyl or represents -C3H7 or -C4H8 optionally substituted in one or more places, the same way or differently with the group -NH2, -NH- (CO)-CH(NH2)-C2H4-COOH, -NH-(CO)-phenyl, -NH-(CO)-CH2- phenyl, -NH-(CO)-CH2-CH(CH3)-phenyl, -NH-(CO)-CH2-O-phenyl, -NH-(CO)O-CH2-phenyl, -NH-(CO)-CH(NH2)CH2-phenyl,
Figure imgf000030_0001
whereby the phenyl can optionally be substituted in one or more places, the same or differently with halogen, -CH3 or -(CO)- C(CH2)(C2H5), or represents C3-alkinyl, as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
In particular the following compounds for general formula (I) are preferred, which inhibit preferentially AKT and/or PDK kinases:
N-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5-bromo- 4-pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide, 1-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5-bromo- 4-pyrimidinyl]amino]propyl]-2-oxo-3-pyrrolidinecarboxylic acid, Λ/-[3-[[5-bromo-4-[[3-[[(5-oxo-2-pyrrolidinyl)carbonyl]amino]propyl]amino]-2- pyrimidinyl]amino]phenyl]-1-pyrrolidinecarboxamide, Pyrrolidine-1 -carboxylic acid [3-(5-bromo-4-{3-[2-(2,4-dichloro-phenyl)- acetylamino]-propylamino}-pyrimidin-2-ylamino)-phenyl]-amide, Pyrrolidine-1 -carboxylic acid [3-(5-bromo-4-{3-[2-(4-bromo-phenyl)-acetylamino]- propylamino}-pyrimidin-2-ylamino)-phenyl]-amide,
Pyrrolidine-1 -carboxylic acid (3-{5-bromo-4-[3-(2-p-tolyl-acetylamino)- propylamino]-pyrimidin-2-ylamino}-phenyl)-amide,
Pyrrolidine-1 -carboxylic acid [3-(5-bromo-4-{3-[2-(2,4-difluoro-phenyl)- acetylamino]-propylamino}-pyrimidin-2-ylamino)-phenyl]-amide, Pyrrolidine-1 -carboxylic acid {3-[5-bromo-4-(3-{2-[2,3-dichloro-4-(2-methylene- butyryl)-phenoxy]-acetylamino}-propylamino)-pyrimidin-2-ylamino]-phenyl}- amide,
Pyrrolidine-1 -carboxylic acid [3-(5-bromo-4-{3-[3-(2,3-dichloro-phenyl)- butyrylamino]-propylamino}-pyrimidin-2-ylamino)-phenyl]-amide, Pyrrolidine-1 -carboxylic acid (3-{5-bromo-4-[3-(3-bromo-benzoylamino)- propylamino]-pyrimidin-2-ylamino}-phenyl)-amide, Λ/-(3-((4-((4-aminobutyl)amino)-5-bromo-2-pyrimidinyl)amino)phenyl)-1- pyrrolidinecarboxamide, N-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5-bromo- 4-pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide,
/^-[3-[[(2S)-2-Amino-1-oxo-3-phenylpropyl]amino]-5-[[5-bromo-4-(prop-2- ynyloxy)pyrimidin-2-yl]amino]phenyl]pyrrolidine-1-carboxamide,
Λ/-[3-[[(2R)-2-Amino-1-oxo-3-phenylpropyl]amino]-5-[[5-bromo-4-(prop-2- ynyloxy)pyrimidin-2-yl]amino]phenyl]pyrrolidine-1-carboxamide,
(αR)-α-Amino-Λ/-[3-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-5-
(hydroxymethyl)phenyl]benzenepropanamide,
2-[3-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-5-hydroxymethyl- phenylaminoj-ethanol,
(2R)-Amino-N-[3-hydroxymethyl-5-(4-prop-2-ynyloxy-pyrimidine-2-ylamino)- phenyl]-3-phenyl-propionamide,
3-((2R)-Amino-3-phenyl-propionylamino)-5-(5-bromo-4-prop-2-ynyloxy- pyrimidine-2-ylamino)- N-cyclopropyl-benzamide, 3-((2R)-Amino-3-phenyl-propionylamino)-5-(5-bromo-4-prop-2-ynyloxy-pyrimidin-
2-ylamino)- N-isopropyl-benzamide,
Phenylmethyl [3-[[2-[[3-[[(ethylamino)carbonyl]amino]phenyl]amino]-5-
(hydroxymethyl)pyrimidine-4-yl]amino]propyl]carbamate,
Pyrrolidine-1 -carboxylic acid (3-{4-[3-((2R)-amino-3-phenyl-propionylamino)- propylamino]-5-bromo-pyrimidine-2-ylamino}-phenyl)-amide,
Pyrrolidine-1 -carboxylic acid (3-{4-[3-((2S)-amino-3-phenyl-propionylamino)- propylamino]-5-bromo-pyrimidine-2-ylamino}-phenyl)-amide,
2-[3-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenylamino]-ethanol,
1-Amino-cyclopentancarbonylic acid[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2- ylamino)-phenyl]-amide,
1-Amino-cyclohexancarbonylic acid-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2- ylamino)-phenyl]-amide,
(2S)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-3- phenyl-propionamide, (2R)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-3- phenyl-propionamide,
2-{[3-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenylamino]-methyl}- phenol,
(2R)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-3-(4- hydroxy-phenyl)-propionamide,
N-[3-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-3-(3,4-dihydroxy- phenyl)-propionamide,
N-[3-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-2-hydroxy-(2S)- phenyl-acetamide,
N-[3-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-2-hydroxy-(2R)- phenyl-acetamide,
(2S)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-3- hydroxy-propionamide,
(2R)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidin-2-ylamino)-phenyl]-3- hydroxy-propionamide,
2-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-2- methyl-propionamide, (2S)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-3-(4- hydroxy-phenyl)-propionamide,
(2S)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-3-p- tolyl-propionamide or
(2R)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-3-p- tolyl-propionamide.
Preferred are also the compounds of general formula (I), which inhibit preferentially AKT and VEGF-R kinases in which A or B in each case independently of one another represent halogen, hydrogen or the group -SO2-CH3, -NO2, -NH2, -CF3, -CH2-NH- (CO)-NH2, -CH2-pyrrolidinyl, -NH-(CO)-CH3, -NH-(CO)-hexyl-NH2, - NH-(CO)-phenyl, -NH-(CO)-pyrrolidinyl, -NH-(CO)-CH(NH2)-CH2- phenyl, NH-(CO)-OCH3, -NH-(CO)-OCH(CH3)2, -NH-(CO)-OC2H4- morpholino, -NH-(CO)-NH-cyclopropyl, -NH-(CO)-morpholino, -NH-
(CO)-NH-C2H4-morpholino, -NH-(CO)-NH-hydroxycycloalkyl, hydantoinyl, whereby the pyrrolidinyl can optionally be substituted with hydroxy or the group -NH2 and whereby the hydantoinyl can optionally be substituted with the group -CH3 or -(CO)-thiazolidinonyl,
X represents the group -NH-,
R1 represents halogen and R2 represents -CH2-dihydroxyphenyl, -C2H -imidazolyl, or -C3H optionally substituted in one or more places, the same way or differently with
Figure imgf000034_0001
as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
In particular the following compounds of general formula (I) are preferred, which inhibit preferrentially AKT and VEGF-R kinases: 4-((4-((2-(1H-imidazol-4-yl)ethyl)amino)-5-iodo-2-pyrimidinyl)amino)- benzenesulfonamide,
N-((3-((5-bromo-4-((2-(1H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)methyl)-urea,
1-((3-((5-bromo-4-((2-(1H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)methyl)-3-pyrrolidinol,
(3-((5-bromo-4-((2-(1H-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)phenyl)- carbamic acid methyl ester,
N2-(3-aminophenyl)-5-bromo-N4-(2-(1 H-imidazol-4-yl)ethyl)-2,4- pyrimidinediamine, N-(3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-N'-cyclopropyl-urea,
N-(3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-4-morpholinecarboxamide,
(3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)phenyl)- carbamic acid 1-methylethyl ester,
N-(3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-methanesulfonamide, N2-(3-amino-5-(trifluoromethyl)phenyl)-5-bromo-N4-(2-(1H-imidazol-4-yl)ethyl)-
2,4-pyrimidinediamine,
N-(3-((5-bromo-4-((2-(1H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-N'-(2-(4-morpholinyl)ethyl)-urea, N2-(3-amino-5-chlorophenyl)-5-bromo-N4-(2-(1 H-imidazol-4-yl)ethyl)-2,4- pyrimidinediamine,
(3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)phenyl)- carbamic acid 2-(4-morpholinyl)ethyl ester,
N-(3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-N'-(4-hydroxycyclohexyl)-urea,
N-(3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-acetamide,
N-(3-((5-bromo-4-((2-(1H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-benzamide, (4R)-N-[3-[[5-bromo-2-[[3-[(1-pyrrolidinylcarbonyl)amino]phenyl]amino]-4- pyrimidinyl]amino]propyl]-2-oxo-4-thiazolidinecarboxamide,
3-[3-[[5-bromo-4-[[2-(1H-imidazol-4-yl)ethyl]amino]-2-pyrimidinyl]amino]phenyl]-
2,4-imidazolidinedione,
3-[3-[[5-bromo-4-[[2-(1 H-imidazol-4-yl)ethyl]amino]-2-pyrimidinyl]amino]phenyl]- 1-methyl-2,4-imidazolidinedione,
1-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5-bromo-
4-pyrimidinyl]amino]propyl]-2-oxo-3-pyrrolidinecarboxylic acid,
1-[3-[[2-[[3-[[(1-aminocyclohexyl)carbonyl]amino]phenyl]amino]-5-bromo-4- pyrimidinyl]amino]propyl]-2-oxo-3-pyrrolidinecarboxylic acid, N-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5-bromo-
4-pyrimidinyl]amino]propyl]-5-oxo-2-pyrrolidinecarboxamide,
N-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5-chloro-
4-pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide,
3-[3-[[5-bromo-4-[[(3,4-dihydroxyphenyl)methyl]amino]-2- pyrimidinyl]amino]phenyl]-2,4-imidazolidinedione,
3-[3-[[5-bromo-4-[[(3,4-dihydroxyphenyl)methyl]amino]-2- pyrimidinyl]amino]phenyl]-1-methyl-2,4-imidazolidinedione,
(4R)-N-[3-[[5-bromo-2-[[3-(2,5-dioxo-1-imidazolidinyl)phenyl]amino]-4- pyrimidinyl]amino]propyl]-2-oxo-4-thiazolidinecarboxamide,
N-[3-[[5-bromo-2-[[3-(2,5-dioxo-1-imidazolidinyl)phenyl]amino]-4- pyrimidinyl]amino]propyl]-5-oxo-2-pyrrolidinecarboxamide,
N-[3-[[5-bromo-2-[[3-(2,5-dioxo-1-imidazolidinyl)phenyl]amino]-4- pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide,
3-[3-[[5-bromo-4-[[3-(2-oxo-1-pyrrolidinyl)propyl]amino]-2- pyrimidinyl]amino]phenyl]-2,4-imidazolidinedione,
(4R)-N-[3-[[5-bromo-2-[[3-(3-methyl-2,5-dioxo-1-imidazolidinyl)phenyl]amino]-4- pyrimidinyl]amino]propyl]-2-oxo-4-thiazolidinecarboxamide or (4R)-N-[3-[[5-bromo-2-[[3-[2,5-dioxo-3-[[(4R)-2-oxo-4-thiazolidinyl]carbonyl]-1- imidazolidinyl]phenyl]amino]-4-pyrimidinyl]amino]propyl]-2-oxo-4- thiazolidinecarboxamide.
It has also been found that compounds of the following structure are inhibitors of kinases, particularly AKT, PDK, Chk, CDK and/ or VEGF-R kinases:
N-(3-((4-((3-(aminomethyl)phenyl)amino)-5-bromo-2-pyrimidinyl)amino)phenyl)-
1 -pyrrolidine-carboxamide,
4-[[5-bromo-4-[[2-(1 H-imidazol-5-yl)ethyl]amino]-2-pyrimidinyl]amino]- 1 - naphthaleneacetic acid, 5-[[5-bromo-4-[[2-(1 H-imidazol-5-yl)ethyl]amino]-2-pyrimidinyl]amino]-1 H-indole-
2-carboxylic acid, ethyl ester,
5-bromo-N4-[2-(1H-imidazol-5-yl)ethyl]-N2-(2-methyl-6-quinolinyl)-2,4- pyrimidinediamine,
4-((5-bromo-4-((2-(" ry-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzamide,
4-((4-((2-( H-imidazol-4-yl)ethyl)amino)-5-iodo-2-pyrimidinyl)amino)- benzenesulfonamide,
3-((5-bromo-4-((2-(7 -/-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzamide, 3-((5-bromo-4-((2-(7/- -imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide,
5-((5-bromo-4-((2-(7/-/-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)-1,3- dihydro-2H-benzimidazol-2-one, 3-((5-bromo-4-((2-( /- -imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzoic acid methyl ester,
3-amino-5-((5-bromo-4-((2-(7H-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzoic acid methyl ester, /V-((3-((5-bromo-4-((2-(W-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)methyl)-methanesulfonamide,
4-((5-bromo-4-((2-(W-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzoic acid methyl ester,
3-((5-bromo-4-((2-( H-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)-phenol, 5-((5-bromo-4-((2-(7H-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- - - isoindole-1 ,3(2H)-dione,
5-bromo-Λ/4-(2-( H-imidazol-4-yl)ethyl)-Λ/2-(3-methylphenyl)-2,4- pyrimidinediamine,
Λ/-(3-((5-bromo-4-((2-( H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-methanesulfonamide,
4-((4-((2-( H-imidazol-4-yl)ethyl)amino)-5-methyl-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((4-((2-(7/-/-imidazol-4-yl)ethyl)amino)-5-(trifluoromethyl)-2-pyrimidinyl)amino)- benzenesulfonamide, 4-((4-((3-aminopropyl)amino)-5-bromo-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((5-bromo-4-((3-( -/-imidazol-1-yl)propyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((5-bromo-4-((2-(1-pyrrolidinyl)ethyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((4-((4-aminobutyl)amino)-5-bromo-2-pyrimidinyl)amino)-benzenesulfonamide,
4-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyrimidinyl)amino)-butanoic acid,
4-((4-((3-((aminocarbonyl)amino)propyl)amino)-5-bromo-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyrimidinyl)amino)-butanoic acid ethyl ester,
4-((5-bromo-4-((4-(methylamino)butyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((5-bromo-4-((2-( /-/-imidazol-1-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((5-ethyl-4-((2-(7H-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((4-((2-(7H-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((5-bromo-4-((2-(2-pyridinyl)ethyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide, 4-((5-bromo-4-((2-(7H-indol-3-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide,
2-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyrimidinyl)amino)- acetamide,
Λ/-(2-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyrimidinyl)amino)ethyl)- acetamide,
3-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyrimidinyl)amino)- propanamide,
Λ/-(4-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyrimidinyl)amino)butyl)- acetamide, Λ/-(3-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyrimidinyl)amino)propyl)- acetamide,
Λ/-(3-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyrimidinyl)amino)propyl)-
2-furancarboxamide,
Λ/-(3-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyrimidinyl)amino)propyl)- H-pyrrole-2-carboxamide,
4-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyrimidinyl)amino)- butanamide,
Λ/-(3-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyrimidinyl)amino)propyl)-
2-thiophenecarboxamide, 4-((4-(4-(aminomethyl)-1-piperidinyl)-5-bromo-2-pyrimidinyl)amino)- benzenesulfonamide,
4-(5-Brom-4-prop-2-ynylamino-pyrimidin-2-ylamino)-phenyl]-N,N- dimethylaminosulfonylamin, 1 -Methyl-1 H-imidazol-4-sulfonsaure [4-(5-brom-4-prop-2-ynylamino-pyrimidin-2- ylamino)-phenyl]-amid,
3-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-benzoic acid ethyl ester, 4-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-benzoic acid ethyl ester, 2-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-benzoic acid ethyl ester, 2-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenol, 4-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-benzoic acid methyl ester, 3-(5-Nitro-4-prop-2-ynylamino-pyrimidine-2-ylamino)-phenol, 2-(5-Nitro-4-prop-2-ynylamino-pyrimidine-2-ylamino)-benzoic acid ethyl ester, 3-(5-Nitro-4-prop-2-ynylamino-pyrimidine-2-ylamino)-benzoic acid ethyl ester, 4-(5-Nitro-4-prop-2-ynylamino-pyrimidine-2-ylamino)-benzoic acid ethyl ester, 4-(5-Nitro-4-prop-2-ynylamino-pyrimidine-2-ylamino)-phenol, Methyl 3-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-5-[(2- hydroxyethyl)amino]benzoate, Methyl 3-amino-5-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]benzoate or 3-[Bis-(2-hydroxy-ethyl)-amino]-5-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2- ylamino)-benzoic acid methyl ester.
Another object of the invention are pharmaceutical composition comprising as an active ingredient at least one compound of general formula (I) or compounds disclosed hereinbefore in an therapeutically effective amount for the prevention or treatment of a disorder caused by, associated with or accompanied by disruptions of cell proliferation and/or angiogenesis together with an pharmaceutically acceptable carrier, diluent or excipient.
A further object of the invention are use of a compound of general formula (I) or compounds disclosed hereinbefore for the manufacture of a medicament for the prevention or treatment of a disorder caused by, associated with or accompanied by any abnormal kinase activity selected from Chk, Akt, Pdk, Cdk and/or VEGF-R activity as well as combinations thereof.
Preferred is the use of compounds of general formula (I), wherein the kinase is selected from PDK1 , Akt1 , Akt2 and/or Akt3, particularly, wherein the kinase is selected from PDK1 , Akt1 , Akt2 and/or Akt3 in combination with VEGF-R or wherein the kinase is selected from Chk1 and/or Chk2.
Another objective of this invention is a method of treating a mammal having a disease-state alleviated by the inhibition of Akt, Pdk, chk and/or VEGF-R activity, wherein the method comprises administering to a mammal a therapeutically effective amount of a compound of general formula (I) or a compound disclosed hereinbefore. In particular the method is objective wherein the mammal is a human.
..Disorders" and/or „disease state,, in the meaning of this invention are selected from cancer, angiofribroma, arthritis, eye diseases, auto-immune diseases, chemotherapy agent-induced alopecia and mucositis, Crohn-disease, endometriosis, fibrotic diseases, hemangioma, cardiovaskular diseases, infectious diseases, nephrological diseases, chronic und acute neurodegenerative diseases, like disruptions of nerval tissue, viral infections, to prevent restenosis of vessels, for preventing the formation of scars, preventing or treating keratoma seniles and contact dermatitis, wherein cancer stands for solide tumours, tumour- or metastasis growth, Kaposis
Sarkom, Hodgkin's disease and/or leukemia, arthritis stands for rheumatoid arthritis, eyes diseases stand for diabetic retinopathy, neovaskular glaukoma, auto-immune diseases stand for psoriasis, alopecia and/or multiple sklerosis, fibrotic diseases stand for cirrhosis of the liver, mesangial cell proliferative diseases, arteriosklerosis, infectiouse diseases stand for diseases that are caused by unicellular parasites, cardiovascular diseases stand for stenosis, like stent induced restenosis, arteriosklerosis and restenosis, nephrological diseases stand for glomerulonephritis, diabetic nephropaty, malignant nephrosklerosis, thrombic mikroangiopathis syndrome, transplant rejections and glomerulopathy, chronic neurodegenerative diseases stand for Huntington's disease, amyotrophic lateralsklerosis, Parkinsons disease, AIDS, dementia und Alzheimer's disease, acute neurodegenerative diseases stand for ischemias of the brain and neurotraumas, and viral infections stand for cytomegalic infections, herpes, hepatitis B or C and HIV.
The compounds according to the invention essentially inhibit on the one hand cell- cycle-associated kinases, particularly serin/threonine kinases, more particularly cyclin-dependent kinases (Cdks), Chks, Akts and/or Pdks or VEGF-R kinases. Preferred is the inhibition of Chks, e.g. Chk1 and/or Chk2, Akts, e.g. Akt1 , Akt2 and/or Akt3 and/or Pdks, e.g. Pdk1.
On the other hand the compounds according to this invention essentially inhibit angiogenesis related kinases, particularly tyrosine kinases, more particularly VEGF-R kinases.
Of particular interest is a preferential inhibition of specific kinases. For example, the compounds of general formula (I) according to claims 2 to 5 show a preferentiality towards Akts, e.g. Akt1 , Akt2 and/or Akt3 and/or Pdks, e.g. Pdk1; the compounds of general formula (I) according to claims 6 to 8 show a preferentiality towards Chks, e.g. Chk1 and/or Chk2 and the compounds of general formula (I) according to claims 9 and 10 show preferentiality towards Akts and VEGF-R kinases upon which is based their action, for example, against cancer, angiofribroma, arthritis, eye diseases, auto-immune diseases, chemotherapy agent-induced alopecia and mucositis, Crohn-disease, endometriosis, fibrotic diseases, hemangioma, cardiovaskular diseases, infectious diseases, nephrological diseases, chronic und acute neurodegenerative diseases, like disruptions of nerval tissue, viral infections, to prevent restenosis of vessels, for preventing the formation of scars, preventing or treating keratoma seniles and contact dermatitis. Compounds of general formula (I) according to claims 9 and 10 show the advantage in the treatment of disorders to have an inhibiting effect of two ways, in particular the cell cycle inhibition and the angiogenesis inhibition due to the preferential inhibition of AKT and VEGF compounds. The eukaryotic cell division ensures the duplication of the genome and its distribution to the daughter cells by passing through a coordinated and regulated sequence of events. The cell cycle is divided into four successive phases: the G1 phase represents the time before the DNA replication, in which the cell grows and is sensitive to external stimuli. In the S phase, the cell replicates its DNA, and in the G2 phase, preparations are made for entry into mitosis. In mitosis (M phase), the replicated DNA separates, and cell division is completed.
The loss of the regulation of the cell cycle and the loss of function of the control points are characteristics of tumor cells.
Changes of the cell cycle control play a role not only in carcinoses. The cell cycle is activated by a number of viruses, both by transforming viruses as well as by non-transforming viruses, to make possible the replication of viruses in the host cell. The false entry into the cell cycle of normally post-mitotic cells is associated with various neurodegenerative diseases. The mechanisms of the cell cycle regulation, their changes in diseases and a number of approaches to develop inhibitors of the cell cycle progression and especially the CDKs were already described in a detailed summary in several publications (Sielecki, T. M. et al. (2000). Cyclin-Dependent Kinase Inhibitors: Useful Targets in Cell Cycle Regulation. J. Med. Chem. 43, 1-18; Fry, D. W. & Garrett, M. D. (2000). Inhibitors of Cyclin-Dependent Kinases as Therapeutic Agents for the Treatment of Cancer. Curr. Opin. Oncol. Endo. Metab. Invest. Drugs 2, 40-59; Rosiania, G. R. & Chang, Y. T. (2000). Targeting Hypeφroliferative Disorders with Cyclin-Dependent Kinase Inhibitors. Exp. Opin. Ther. Patents 10, 215-230; Meijer L. et al. (1999). Properties and Potential Applications of Chemical Inhibitors of Cyclin-Dependent Kinases. Pharmacol. Ther. 82, 279-284; Senderowicz, A. M. & Sausville, E. A. (2000). Preclinical and Clinical Development of Cyclin-Dependent Kinase Modulators. J. Natl. Cancer Inst. 92, 376-387).
The pivotal role of VEGF and of its receptors during vascular development was exemplified in studies on targeted gene inactivation. Even the heterozygous disruption of the VEGF gene resulted in fatal deficiencies in vascularization (Carmeliet et al., Nature 380, 435-439, 1996; Ferrara et al., Nature 380, 439- 442, 1996). Mice carrying homozygous disruptions in either Flt1 or Flk1/KDR gene die in mid-gestation of acute vascular defects. However, the phenotypes are distinct in that Flk1/KDR knock-out mice lack both endothelial cells and a developing hematopoietic system (Shalaby et al. Nature 376, 62-66, 1995), whereas Flt1 deficient mice have normal hematopoietic progenitors and endothelial cells, which fail to assemble into functional vessels (Fong et al., 376, 66-70, 1995). Disruption of the Flt4 gene, whose extensive embryonic expression becomes restricted to lymphatic vessels in adults, revealed an essential role of Flt4 for the remodeling and maturation of the primary vascular networks into larger blood vessels during early development of the cardiovascular system (Dumont et al., Science 282, 946-949, 1998). Consistent with the lymphatic expression of Flt4 in adults overexpression of VEGF-C in the skin of transgenic mice resulted in lymphatic, but not vascular, endothelial proliferation and vessel enlargement (Jeltsch et al., Science 276, 1423-1425, 1997). Moreover, VEGF-C was reported to induce neovascularization in mouse cornea and chicken embryo chorioallantoic membrane models of angiogenesis (Cao et al., Proc. Natl. Acad. Sci. USA 95, 14389-14394, 1998).
In pathological settings associated with aberrant neovascularization elevated expression of angiogenic growth factors and of their receptors has been observed. Most solid tumors express high levels of VEGF and the VEGF receptors appear predominantly in endothelial cells of vessels surrounding or penetrating the malignant tissue (Plate et al., Cancer Res. 53, 5822-5827, 1993). Interference with the VEGF/VEGF receptor system by means of VEGF- neutralizing antibodies (Kim et al., Nature 362, 841-844, 1993), retroviral expression of dominant negative VEGF receptor variants (Millauer et al., Nature 367, 576-579, 1994), recombinant VEGF-neutralizing receptor variants (Goldman et al., Proc. Natl. Acad. Sci. USA 95, 8795-8800, 1998), or small molecule inhibitors of VEGF receptor tyrosine kinase (Fong et al., Cancer Res. 59, 99-106, 1999; Wedge et al., Cancer Res. 60, 970-975, 2000; Wood et al. Cancer Res. 60, 2178-2189, 2000), or targeting cytotoxic agents via the VEGF/VEGF receptor system (Arora et al., Cancer Res. 59, 183-188, 1999; EP 0696456A2) resulted in reduced tumor growth and tumor vascularization. However, although many tumors were inhibited by interference with the VEGF/VEGF receptor system, others were unaffected (Millauer et al., Cancer Res. 56, 1615-1620, 1996). Human tumors as well as experimental tumor xenografts contain a large number of immature blood vessels that have not yet recruited periendothelial cells. The fraction of immature vessels is in the range of 40% in slow growing prostate cancer and 90% in fast growing glioblastoma. A selective obliteration of immature tumor vessels was observed upon withdrawal of VEGF by means of downregulation of VEGF transgene expression in a C6 glioblastoma xenograft model. This result is in accordance with a function of VEGF as endothelial cell survival factor. Similarly, in human prostate cancer shutting off VEGF expression as a consequence of androgen-ablation therapy led to selective apoptotic death of endothelial cells in vessels lacking periendothelial cell coverage. In contrast, the fraction of vessels which resisted VEGF withdrawal showed periendothelial cell coverage (Benjamin et al., J. Clin. Invest. 103, 159-165, 1999).
To use the compounds according to the invention as pharmaceutical agents, the latter are brought into the form of a pharmaceutical preparation, which in addition to the active ingredient for enteral or parenteral administration contains suitable pharmaceutical, organic or inorganic inert carrier materials, such as, for example, water, gelatin, gum arabic, lactose, starch, magnesium stearate, talc, vegetable oils, polyalkylene glycols, etc. The pharmaceutical preparations can be present in solid form, for example as tablets, coated tablets, suppositories, or capsules, or in liquid form, for example as solutions, suspensions, or emulsions. Moreover, they optionally contain adjuvants, such as preservatives, stabilizers, wetting agents or emulsifiers; salts for changing the osmotic pressure or buffers. These pharmaceutical preparations are also subjects of this invention.
For parenteral administration, especially injection solutions or suspensions, especially aqueous solutions of active compounds in polyhydroxy-ethoxylated castor oil, are suitable. As carrier systems, surface-active adjuvants such as salts of gallic acids or animal or plant phospholipids, as well as mixtures thereof and liposomes or ingredients thereof can also be used.
For oral administration, especially tablets, coated tablets, pills or capsules with talcum and/or hydrocarbon carriers or binders, such as, for example, lactose, maize or potato starch, are suitable. The oral application can also be in a liquid form, such as, for example, as a juice, to which optionally a sweetener is added.
Enteral, parenteral and oral administrations are also subjects of this invention. The dosage of the active ingredients can vary depending on the method of administration, age and weight of the patient, type and severity of the disease to be treated and similar factors. The daily dose is 0.5-1000 mg, preferably 50-200 mg, whereby the dose can be given as a single dose to be administered once or divided into two or more daily doses.
If the production of the starting compounds for the manufacture of the compounds of the invention is not described, these starting compounds are known or can be produced analogously to known compounds or to processes that are described here. It is also possible to perform all reactions that are described here in parallel reactors or by means of combinatory operating procedures.
The isomer mixtures can be separated into the enantiomers or E/Z isomers according to commonly used methods, such as, for example, crystallization, chromatography or salt formation.
The production of the salts is carried out in the usual way by a solution of the compound of formulae l-VII being mixed with the equivalent amount of or excess base or acid, which optionally is in solution, and the precipitate being separated or the solution being worked up in the usual way. Inhibition of Pdk/Akt activity
General remarks
Compounds described herein, potently block an assay in which phosphoinositide-dependent kinase-1 (PDK-1) mediates the activation of AKT, whose activity is measured in the assay. The compounds, therefore, can be blocking the assay by inhibiting PDK-1 enzyme activity, AKT enzyme activity, or the activation of AKT by PDK-1. These compounds are expected to be therapeutically useful in cancer by inhibiting processes critical for tumor progression, including cell proliferation, survival, and tumor angiogenesis (Testa and Bellacosa 2001; Vivanco and Sawyers 2002). As described herein, compounds blocking block colony formation and/or growth of PC-3 prostate and MDA-468 breast cancer cells in soft agar, which is an in vitro measure of potential anti-tumor activity. Furthermore, the compounds described herein are expected to sensitize tumors to the effects of other chemotherapeutic agents and radiation (Page, Lin et al. 2000; Brognard, Clark et al. 2001).
PDK-1 is a Ser/Thr kinase that functions to phosphorylate and activate other Ser/Thr kinases in the AGC kinase family (Vanhaesebroeck and Alessi 2000).
The best-characterized substrate of PDK-1 is the intracellular Serine/Threonine kinase AKT, whose expression and/or activity is elevated in many cancers.
Kinase activity of serum and glucocordicoid regulated kinase (SGK), which is structurally related to AKT, can also be phosphorylated and activated by PDK-1. Once activated in tumors, AKT promotes increase tumor cell survival, drug resistance, growth and angiogenesis. Three highly related isoforms of AKT, termed AKT1 , AKT2 and AKT3 are known in humans. Activation of AKT is dependent on the activity of phosphatidylinsoitol-3 kinase (PI-3 kinase), whose activity is activated by many signaling molecules elevated in cancer cells, including growth factor receptors (e.g., epidermal growth factor (EGF) receptor,
ErbB2 and IGF 1 -receptor) and oncogenes (e.g, Ras, BCR-abl, and Src). Other potential substrates of PDK-1 include p70 S6 kinase, p90 S6 kinase, protein kinase C, cAMP-dependent protein kinase (PKA), PRK1 , Protein kinase G and serum and glucocorticoid regulated kinase (SGK).
PDK-1 -mediated phosphorylation of AKT, which is largely in an inactive form in unstimulated cells, converts the enzyme to a catalytically active form. This occurs through the phosphorylation of the activation loop domain of AKT e.g., at Threonine-309 in AKT2 and Theonine-308 in AKT1. Phosphorylation of a homologous domain in many kinases is known to regulate their kinase activity. One stimulus for PDK-1 mediated phosphorylation of AKT is the association PI-3 kinase products (3,4,5)PIP3 or (3,4)PIP2 with the pleckstrin homology (PH) domain of AKT. Although AKT displays low, basal levels of activation in normal, unstimulated cells, AKT often becomes constitutively activated in tumor cells. This occurs through the up-regulation of a variety of different signaling molecules or the presence of oncogenenic mutations commonly found in cancer cells that can promote the activation of AKT, such as PI-3 kinase, growth factor receptors (e.g., EGFR family members), Ras, Src, and BCR-ABL activation. Loss of the tumor suppressor PTEN is another means of greatly increasing AKT activity in cancer cells (Besson, Robbins et al. 1999). PTEN mutation or down regulation of PTEN protein is found in a large number of tumors and cancer cell lines. PTEN is a phosphatase that removes the D-3 phosphate from the products of PI-3 kinase such as phosphatidylinositol 3,4,5-trisphosphate and phosphatidylinositol 3,4-bisphosphate (Myers, Pass et al. 1998; Stambolic, Suzuki et al. 1998). Loss of PTEN, therefore, has the effect of increasing products of PI-3 kinase and promoting constitutive activation of AKT. Cancers with highly up-regulated levels of AKT may be especially sensitive to the effects of PDK-1 /AKT pathway inhibitors.
Downstream substrates of PDK-1 and/or AKT are associated with a number of cell responses including proliferation, metabolism and cell survival (Testa and Bellacosa 2001; Vivanco and Sawyers 2002). Examples of signaling molecules downstream from PDK-1 or AKT involved in these pathways include BAD, p70 S6 kinase, p21(Waf-1/Cip-1), Forkhead transcription factors, p27(kip-1), GSK-3-alpha/beta, TSC2 (tuberin), and ecNOS. The survival function of AKT is particularly well-characterized cellular activity of AKT (Datta, Brunet et al. 1999). AKT functions to suppress apoptosis induced by a variety of agents, including UV radiation, chemotherateutic drugs, TFG-beta, withdrawal of survival factors, overexpression of oncogenes such as c-myc and detachment of cells from the extracellular matrix.
The ability to escape cell death, also termed apoptosis, is critical characteristic of tumor cells allowing their uncontrolled growth and invasive behavior. One trigger for apoptosis is the perturbation of the normal growth regulation resulting from oncogenic mutations or inappropriate expression signaling molecules coupled to cell proliferation. Apoptotic pathways, therefore, provide a key means of protection from the development and progression of cancer. Cancer cells, however, can escape apoptotic death by selecting for activation of signaling molecules such as AKT that turn off apoptotic signals. Some oncogenes, such as Ras, which is activated in as many as 60% of human tumors, simultaneously promote uncontrolled growth and the activation of AKT. Inhibition of AKT in HIH 3T3 cells prevents transformation of these cells through transfection with activated Ras. Furthermore, a number of studies have shown that combining expression an oncogene with an activated form of AKT greatly facilitates formation of tumors in vivo (e.g., (Holland, Celestino et al. 2000)). Inhibitors of PDK-1, by blocking activation of AKT, are a means of promoting apoptosis in tumors cells, especially, but not necessarily limited to those over-expressing AKT activity. By blocking cell survival mechanisms, the compounds described herein could also be useful to promote sensitivity of cancer cells to radiation therapy and to treatment with a variety of chemotherapeutic agents.
Inhibitors of the PDK-1 /AKT pathway are also expected to block cancer progression through inhibition of tumor-stimulated angiogenesis (Dimmeler and Zeiher 2000; Shiojima and Walsh 2002). AKT has been shown to regulate a number of responses critical for the process of angiogeneisis, including endothelial cell migration, proliferation and survival during new vessel formation, ecNOS regulation, response of endothelial cells to growth factors (including IGF-1 , agniopoetin-1 and VEGF) and the regulation of hypoxia-inducible factor-1 (HIF-l)-alpha levels.
Inhibition of the cell cycle and growth of tumor cells is yet another expected effect of compounds that block PDK-1 and/or AKT. Inhibition of PDK-1 and/or AKT activity has been shown to regulate growth of cancer cells in a number of studies. These effects may occur through PDK-1 or AKT-mediated regulation of a number of different signaling pathways important in growth regulation. For example, AKT has been shown to block nuclear localization and/or expression of the cyclin-dependent kinase inhibitors, p21(Waf-1/Cip-1) and p27(kip-1). Compounds blocking these effects would be expected to reduce the activity of cyclin-dependent kinases, blocking progression through the cell cycle and reducing tumor cell growth. AKT was found to inhibit Myt1 , thereby acting as an initiator of mitosis in oocytes fronm the starfish Asterina pectinfera. Furthermore, PDK-1 and/or AKT regulate the expression of proteins important for cell growth through its regulation of mTOR, p70 S6 kinase and eukaryotic initiation factor 4E binding protein 1 (4E-BP1). While the mechanism of this regulation is not firmly established, it has been shown that AKT phosphorylations and reduces expression of TSC2, thereby relieving TSC-2 mediated suppression of mTOR activity. This, in turn, promotes the activation p70 S6 kinase activity and the phosphorylation and inhibition of 4E-BP1 (Inoki, Li et al. 2002; Potter, Pedraza et al. 2002). Both these effects result in increased synthesis of mRNAs encoding proteins important for cell growth. Loss of TSC2 function is associated with the disease tuberous sclerosis, which results in differentiated benign growths (harmatomas) in a wide variety of organs. PDK-1 also has been shown to have a direct role in the phosphorylation and activation p70 S6 kinase (Alessi, Kozlowski et al. 1998).
In summary, the compounds described which block PDK-1 mediated activation of AKT or PDK-1 directly may be useful therapeutic agents in cancer by blocking a number of processes required for tumor progression, including growth, tumor cell survival, and recruitment of new blood vessels. The compounds described may also enhance the anti-tumor effects of radiation or other chemotherepeutic drugs. The compounds may also be useful for the treatment of tuberous sclerosis. Furthermore, the compounds described could be useful modulators of the immune response (Cantrell 2002) and for the treatment of autoimmune diseases such as rheumatoid arthritis and MS.
Experimental Procedures 1
Cell-based assays
Materials: Prostate cancer cells (PC-3) and breast cancer cells (MDA-468) were obtained from the ATCC (Manassas, VA). Mammalian protein extraction reagent (MPER), Halt protease inhibitor cocktail, BCA protein reagent, and Supersignal Western Chemiluminescent reagent were obtained from Pierce Chemical Co. (Rockford, IL). 10% Tris-Glycine gels (1.0mm, 15-well) and nitrocellulose (0.2 micron) were obtained from Invitrogen Life Technologies (Carlsbad, CA). Agar agar was purchased from EM Science. Polyclonal antibodies raised against phospho-AKT (Thr308, #9275), phospho-AKT (Ser473, #9271), phospho-S6-kinase (Thr389, #9205), and anti-rabbit IgG-HRP conjugate were obtained from Cell Signaling Technologies (Beverly, MA). Nitroblue tetrazolium reagent and staurosporine were purchased from Sigma Chemical Co. (St. Louis, MO). LY294002 was purchased from Cayman Chemicals (Ann Arbor, Ml). All other materials were of reagent-grade quality.
Cell growth conditions: PC-3 cells were grown in F12K medium, supplemented with 7% (v/v) fetal calf serum (fcs) and 2mM glutamine. MDA-468 cells were grown in MEM-alpha, supplemented with 10% (v/v) fcs, 2mM glutamine, 1mM sodium pyruvate, 0.1 mM non-essential amino acids, 10mM Hepes, and 1 μg/ml insulin. All cell lines were incubated in a 37DC humidified incubator, with a 5% CO2 atmosphere.
Cell-based assays using Western blot analysis: PC-3 cells were seeded into 24-well plates (Corning Costar) at 100-120,000 cells per well and allowed to grow overnight to 90% confluence. On the next day, the cells were washed once with 1.5ml PBS, and the medium replaced with low serum (0.1% fcs) containing growth medium (starvation medium). After a second overnight incubation, the medium was replaced with 0.5ml/well of starvation medium. Some assays were also conducted in normal growth medium (7% fcs, PC-3, or 10% fcs, MDA-468). Cells were treated with vehicle control (DMSO) or drug at a final DMSO concentration of 1% v/v (a 5μl addition per 0.5ml medium), and cells were allowed to incubate for the stated times. The incubations were terminated by aspiration of the medium, washing the wells with 1.0ml PBS, and lysis in 0.1ml MPER reagent, supplemented with protease inhibitors (Halt reagent) and phosphatase inhibitors (1mM NaF, 1mM sodium vanadate). Cell lysates were briefly centrifuged to remove insoluble debris, and aliquots were taken for protein (BCA) and Western blot analysis. For Western analysis, lysates were combined with Laemmli SDS sample buffer, boiled, and loaded onto 10% Tris-Glylcine gels, normalizing for the amount of protein loaded in each lane. Electrophoresed gels were transferred onto nitrocellulose paper, blocked with 5% milk in Tris-buffered saline containing 0.1% Tween-20, and incubated overnight with the primary antibody (phospho-AKT-Thr308 @ 1 :667, phospho-AKT-Ser473 @ 1:1000, phospho-S6 kinase @ 1 :1000). Blots were washed three times with blocking buffer and incubated one hour with anti-rabbit IgG-HRP @ 1:2000. Washed blots were developed using the Supersignal Western Chemiluminescent detection system. Films were scanned using a Bio Rad CCD camera, and phospho-protein bands were quantitated using Bio Rad Quantity-One software.
Soft agar efficacy assays: PC-3 and MDA-468 cells were grown in soft agar over a period of 2 weeks. Culture plates (Corning 35mm x 10mm) were prepared with a bottom layer of 0.5% agar in growth medium, 2ml/well. Cells were trypsinized, dispersed into single cells with a 21 -gauge needle, and seeded in a top layer of 0.3% agar/growth medium, 1.5ml/plate, containing 4500 cells per plate. On the following day, the first vehicle or drug treatment was added, in a volume of 1.0ml of 0.3% agar/growth medium, containing 1% DMSO. Drug concentrations were adjusted to reflect the total volume of agar in the plates. The cells were allowed to grow for seven days and treated a second time (adding an additional 1 ml of 0.3% agar). Colonies were visually inspected for growth and viability every few days. On day 12-14, nitroblue tetrazolium (0.5 mg/ml PBS) was added, 0.3 ml per plate, and the viable colonies were allowed to develop color for 1-2 days. Plates were scanned using a Bio Rad CCD camera, and the colonies were quantitated for ony number, and for total stained area, using ImagePro software.
AKT2 and PDK-1 Expression and purification
pHisAKT2 was constructed by cloning AKT2 into pBlueBacHis2A (Invitrogen Corp.) through the BamH1 and Bgl2 restriction sites, forming a fusion protein behind a 38 amino acid N-terminal His tag sequence derived from the vector. The new N-terminal sequence + first 10 residues of AKT2 is as follows: MPRGSHHHHHHGMASMTGGQQMGRDLYDDDDKDRWGSMNEVSVIKEG (AKT2 is underlined and is in bold His-6). Similarly, pHisPDK-1 was constructed by cloning PDK1 into pBlueBacHis2A (Invitrogen Coφ.) at EcoR1 cloning site, forming a fusion protein behind an N-terminal His-tag (preceding sequence of ...ICSWYHGILDMARTTSQLYD.... (PDK1 sequence underlined). The new N-terminal sequence + first 10 residues of PDK1 is as follows: MPRGSHHHHHHGMASMTGGQQMGRDLYDDDDKDRWGSELEICSWYHGILD MARTTSQLYD- (PDK1 is underlined and His-6 is in bold).
Recombinant baculovirus containing either His-tagged AKT2 or His-tagged PDK-1 cDNAs were prepared by the following method. pHisAKT2 or pHisPDK-1 were cotransfected with Bac-N-Blue (Invitrogen) viral DNA info SF-21 cells and after 3 - 4 days, viral supernatant were isolated and recombinant viruses were plaque purified. His-tagged AKT2 (HisAKT-V) or His-tagged PDK-1 (HisPDK-1-V) cDNA expressing clones were selected and expanded as a stock for use in the expression of recombinant proteins described below.
To express His-tagged AKT2 and PDK-1, a 10 liter suspensions of SF-21 insect cells were infected with recombinant viruses (i.e., either HisPDK-1-V or HisAKT2-V) and cells were harvested 3-4 days post infection and frozen. To purify recombinant His-tagged AKT2 and PDK-1, cell pellets were thawed, homogenized (in phosphate buffered saline (PBS), supplemented with 10% Triton X-100, 0.5 M NaCl, 2 g/l NaF, 2.5 μg/ml aprotinin, 5 μg/ml leupeptin, 1.25 μg/ml pepstatin, 0.1% beta-mecaptoethanol, and 1 mM vanidate, 10 mM imidizole and adjusted to pH 7.6) and were purified using two sequential rounds of Ni2+ affinity chromatography followed by gel filtration. Enzymes were frozen in small aliquots and stored at -80DC in 50 mM Tris-HCl, pH 7.5, 150 mM NaCl, pH 7.5, 0.1 mM EGTA, 0.1 mM EDTA, 0.2 μM benzamidine, 0.1% beta-mercaptoethanol and 0.25 M sucrose.
Enzyme Assays
PDK-1 -dependent activation and subsequent enzymatic activity of AKT2: Purified human AKT2 activity was routinely measured in an assay in which the enzyme was first activated by PDK-1 in the presence of phosphatidylinositol-4,5-bisphosphate (PIP2). Once activated, AKT2-dependent phosphorylation of a peptide substrate was measured by scintillation proximity assay (SPA).
Phospholipid vesicles were prepared as follows: 2.2 mg each of phosphatidylcholine (Sigma Cat # P-1287) and phosphatidylserine (Sigma Cat #P-6641) were transferred to a borosilicate glass test tube and dried down under nitrogen. 1 mg of PIP2 (Biomol Cat #PH-106) was suspended in 9.5 ml of 10 mM HEPES, pH 7.5 and transferred to the dried lipids. The tube was vortexed until a milky suspension was produced. Then the tube was placed in a ice water-jacketed cup horn sonicator (Branson Instruments) and subjected to sonication for 20 min at medium power until a translucent phospholipid vesicle preparation was obtained. Aliquots of the vesicle suspension were frozen at -80DC until needed.
Assays were performed in 96-well polypropylene V-bottom plates. Incubations were carried out for 2 hr at room temperature. The assay mixture contained in a volume of 60μL: 15 mM MOPS, pH 7.2, 1 mg/ml bovine serum albumin, 18 mM betaglycerolphosphate, 0.7 mM dithiothreitol, 3 mM EGTA, 10 mM MgOAc, 7.5 (M ATP, 0.2 μCi of [γ-33P]ATP, 7.5 μM biotinylated peptide substrate (biotin-ARRRDGGGAQPFRPRAATF), 0.5 μL of PIP2-containing phospholipid vesicles, 60 pg of purified recombinant human PDK-1 , and 172 ng of purified recombinant human AKT2. Test compounds were added from stock solutions in DMSO. The final concentration of DMSO was 2.5%. Following incubation, 10 μL of the assay mixture was transferred to a 96-well clear-bottom polystyrene plate (Wallac Isoplate) containing 0.33 mg of streptavidin-coated SPA beads (Amersham Cat. # RPNQ0007) suspended in 200 μL of phosphate-buffered saline, pH 7.4, containing 50 mM EDTA and 0.1% Triton X-100. After brief shaking, the SPA beads were allowed to settle to the bottom of the plate overnight at room temperature. Product formation, measured in a Wallac MicroBeta scintillation counter, was proportional to the time of incubation and to the amount of PDK-1 and inactive AKT2 added. PDK-1 was added at sub-optimal levels so that the assay could sensitively detect inhibitors of AKT2 activation as well as direct AKT2 kinase inhibitors. The z'-factor for the assay was greater than 0.7.
Phosphorylation of the peptide substrate on the threonine residue was shown to be dependent upon activated AKT2 produced during the incubation. No phosphorylation was observed in the absence of ATP, Mg2+, PDK-1 , AKT2, or PIP2-containing vesicles. Phosphorylation was readily observed, however, upon addition of purified activated human AKT1 (purchased from Upstate Biotechnology), independent of the presence or absence of added PDK-1 or PIP2-containing vesicles.
Direct assay of PDK-1 activity: Recombinant human PDK-1 activity was directly measured using a filter binding protocol. Incubations were performed at room temperature for 4 hr in a final volume of 60 μL containing: 50 mM Tris-HCl, pH 7.5, 0.1 mM EGTA, 0.1 mM EDTA, 0.1% beta-mercaptoethanol, 1 mg/ml bovine serum albumin, 10 mM MgOAc, 10 μM ATP, 0.2 μCi of [γ-33P]ATP, 7.5 μM of substrate peptide (H2N-ARRRGVTTKTFCGT) and 60 ng of purified human PDK-1. The enzymatic reaction was stopped by addition of 25 mM EDTA. A portion of the reaction mixture was spotted on Whatman P81 phosphocellulose paper. The filter paper was washed 3 times with 0.75% phosphoric acid to remove unreacted [γ-33P]ATP, and once with acetone. After drying, the filter-bound labeled peptide was quantitated using a Fuji Phosphoimager. Results
Compounds, which preferentially inhibit Akt/Pdk activity are shown in figure 1.
An overview of the results of the inhibition IC5o in nM are presented in the table 1 below:
Table 1:
Figure imgf000055_0001
References:
Alessi, D. R., M. T. Kozlowski, et al. (1998). "3-Phosphoinositide-dependent protein kinase 1 (PDK1) phosphorylates and activates the p70 S6 kinase in vivo and in vitro." Curr Biol 8(2): 69-81.
Besson, A., S. M. Robbins, et al. (1999). "PTEN/MMAC1/TEP1 in signal transduction and tumorigenesis." Eur J Biochem 263(3): 605-11.
Brognard, J., A. S. Clark, et al. (2001). "Akt/protein kinase B is constitutively active in non-small cell lung cancer cells and promotes cellular survival and resistance to chemotherapy and radiation." Cancer Res 61(10): 3986-97.
Cantrell, D. (2002). "Protein kinase B (Akt) regulation and function in T lymphocytes." Semin Immunol 14(1): 19-26.
Datta, S. R., A. Brunet, et al. (1999). "Cellular survival: a play in three Akts." Genes Dev 13(22): 2905-27. Dimmeler, S. and A. M. Zeiher (2000). "Akt takes center stage in angiogenesis signaling." Circ Res 86(1): 4-5.
Holland, E. C, J. Celestino, et al. (2000). "Combined activation of Ras and Akt in neural progenitors induces glioblastoma formation in mice." Nat Genet 25(1): 55-7.
Inoki, K., Y. Li, et al. (2002). "TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling." Nat Cell Biol 12: 12.
Myers, M. P., I. Pass, et al. (1998). "The lipid phosphatase activity of PTEN is critical for its tumor supressor function." Proc Natl Acad Sci U S A 95(23): 13513-8.
Page, C, H. J. Lin, et al. (2000). "Overexpression of Akt/AKT can modulate chemotherapy-induced apoptosis." Anticancer Res 20(1 A): 407-16.
Potter, C. J., L. G. Pedraza, et al. (2002). "Akt regulates growth by directly phosphorylating Tsc2." Nat Cell Biol 12: 12. Shiojima, I. and K. Walsh (2002). "Role of Akt signaling in vascular homeostasis and angiogenesis." Circ Res 90(12): 1243-50.
Stambolic, V., A. Suzuki, et al. (1998). "Negative regulation of PKB/Akt-dependent cell survival by the tumor suppressor PTEN." Cell 95(1): 29-39.
Testa, J. R. and A. Bellacosa (2001). "AKT plays a central role in tumorigenesis." Proc Natl Acad Sci U S A 98(20): 10983-5.
Vanhaesebroeck, B. and D. R. Alessi (2000). "The PI3K-PDK1 connection: more than just a road to PKB." Biochem J 346(Pt 3): 561-76.
Vivanco, I. and C. L. Sawyers (2002). "The phosphatidylinositol 3-Kinase AKT pathway in human cancer." Nat Rev Cancer 2(7): 489-501.
Inhibition of Chk kinase activity
General Remarks
The compounds of this invention inhibit the cell cycle checkpoint kinases which are essential for the cellular response to DNA damage and for the coordination of the cell cycle. The DNA damage might be due to external or internal influence. These influences involve - without being limited to them - replication errors, DNA base damages, DNA strand breaks and the exposition to irradiation or cytotoxic chemicals.
The inhibition of one or more of the cell cycle checkpoint kinases is the basis for the effect of the compounds of this invention e.g. against cancer, like solid tumours or leukemia, against other hyperproliferative diseases, e.g. HIV and viral infections, like e.g. cytomegalus-infections, herpes and hepatitis B and C and HIV.
The eukaryotic cell division cycle ensures the duplication of the genome and its correct distribution to the daughter cells by running through a coordinated and regulated sequence of events. The cell cycle is divided in four successive phases: the G1 phase represents the time before the DNA replication, during which the cell is growing and susceptible for external stimuli. During the S-phase the cell replicates its DNA, and in the G2 phase the cell prepares for the entry into the mitosis. During the mitosis (M-Phase) the replicated DNA is separated and the cell division is carried out.
Corresponding to the extraordinary relevance of the cell division cycle the passage through the cycle is strictly regulated and controlled. The enzymes needed for the progression through the cycle, the cyclin-dependent kinases, have to be activated at the right moment and have to be switched off as soon as the corresponding phase is finished. Checkpoint systems arrest the progression through the cell cycle if DNA damage is detected, the DNA replication is not completed or the building of the spindel apparatus is not completed (Hartwell et al., 1989). They do this by influencing the generation, activation or inactivation of the cyclin-dependent kinases.
Checkpoints permit the cell to track the ordered course of the individual phases of the cell cycle. The most important checkpoints are at the transition from the G1 phase into the S phase and at the transition from the G2 phase into the M phase
(for a review see Dasika et al. 1999). The G1 checkpoint ensures that the cell does not start the DNA synthesis if it is not sufficiently nourished or if it does not correctly interact with other cells or with the substrate or if the DNA of the cell is not intact. The G2/M checkpoint ensures that the DNA is completely replicated and the mitotic spindle is build up before the cell enters the mitosis. The G1 checkpoint is controlled by the gene product of the tumour suppressor gene p53. p53 becomes activated after the detection of changes in the metabolism or the genomic integrity of the cell and p53 is able to initiate either a stop of the cell cycle program or apoptosis. For this the transcriptional activation of the expression CDK inhibiting protein p21 plays a crucial role.
A fundamental component of the G2/M checkpoint is the activation of the kinases ATM, Chk1 and Chk2 after a DNA damage and finally the phosphorylation and inactivation of the phosphatase Cdc25C. This results in a cell cycle arrest, as the inhibitory phosphorylation of the amino acids threonine-14 and tyrosine-15 of the cyclin dependent kinase 1 (CDK1) is not further removed by Cdc25C.
The loss of the regulation of the cell cycle and the loss of checkpoint control are characteristic features of tumour cells. p53, which is essential for the G1 checkpoint, is the gene most often mutated in human tumours (about 50 %). In tumour cells expressing unmutated p53, it is often inactivated by an enhanced proteolytic degradation or the genes of other proteins involved in the G1 checkpoint are mutated or deregulated. Examples are the inactivation of the tumour suppressor genes Rb, p16INK4 and p19ARF or the overexpression of the oncogenes HDM-2 and cyclin D (Levine, 1997). In consequence nearly all tumour cells do not have a functional G1 checkpoint which enables the to accumulate further mutations and to escape from a DNA damage induced apoptosis. This inactivation of the G1 checkpoint is an important factor for the genomic instability which drives the evolution of human tumours and crucially contributes to the resistance of tumour cells against chemotherapeutics and irradiation. On the other hand the inactivation of the G1 checkpoint enhances the dependence of the tumour cells on the second important barrier against the cell killing effect of DNA damages, the G2/M checkpoint, and makes the tumour cells especially vulnerable to an abrogation of the G2/M checkpoint (Hartwell und Kastan, 1994, O'Connor und Fan, 1996).
The cell cycle checkpoint kinase Chk1 is an important part of the G2/M checkpoint (Sanchez et al., 1997). Inactivation of Chk1 abrogates a DNA damage induced G2/M arrest and thereby leads to a preferred killing of the resulting checkpoint deficient cells (Takai et al., 2000, Koniaras et al., 2001 , Liu et al., 2000). The inactivation of Chk1 causes that Cdc25C stays active despite of the DNA damage and is able to activate Cdk1/CycB, the main effector of the entry into the mitosis. However, due to the persistent DNA damage the cell is not able to complete the M phase successfully and undergoes apoptosis instead ("mitotic catastrophe"). The cell cycle checkpoint kinase Chk2 is also activated by DNA damage (Matsuoka et al. 1998, Chaturvedi et al., 1999) and activated Chk2 phosphorylates and thereby inactivates Cdc25C. Cells without active Chk2 have a defect in their checkpoint response to DNA damage (Hirao et al., 2000). The inactivation of Chk1 and Chk2 abrogates the G2/M arrest which is induced by damaged DNA and sensitises the resulting checkpoint deficient cells to the killing by DNA damaging events. As cancer cells are more sensitive towards the abrogation of the G2/M checkpoint than normal cells there is great interest in compounds, which inhibit Chk1, Chk2 or Chk1 and Chk2, as a result abrogate the G2/M checkpoint and improve the killing of cancer cells by DNA damaging events. Such DNA damaging events can be the direct damage of the DNA by irradiation or chemotherapeutics, e.g. strandbreaks inducing compounds, DNA-alkylating compounds or topoisomerase inhibitors, the exertion of influence on the building of the mitotic spindle apparatus, hypoxic stress due to limited supply of the tumour with blood - e.g. induced by anti-angiogenic drugs - or also endogenous DNA damages resulting from the genomic instability inherent to cancer cells. Experimental Procedure 2
Chk1 kinase assay
Recombinant Chk1-His6-fusion protein, expressed in insect cells (Sf-9) and purified by Ni-NTA affinity chromatography was used as kinase. Alternatively, commercially available GST-Chk1-fusion protein (Upstate Biotechnology, Dundee, Scotland) can be used. As substrate for the kinase reaction the biotinylated peptide biotin-Arg-Ser-Gly-Leu-Tyr-Arg-Ser-Pro-Ser-Met-Pro-Glu-Asn-Leu-Asn-Arg-Pro- Arg-OH was used which can be purchased e.g. from the company Biosyntan GmbH (Berlin-Buch, Germany).
Chk1 (200 ng/measurement point) was incubated for 60 min at 22 DC in the presence of different concentrations of test compounds (0 μM and concentrations in the range 0.001 - 30 μM) in 30 μl assay buffer [50 mM Hepes/NaOH pH7.5, 10 mM MgCI2, 1 mM MnCI2, 0.1 mM sodium ortho-vanadate, 1.0 mM dithiothreitol, 0.5 μM adenosine-tri-phosphate (ATP), 1.9 μM substrate peptide
(Biotin-Arg-Ser-Gly-Leu-Tyr-Arg-Ser-Pro-Ser-Met-Pro-Glu-Asn-Leu-Asn- Arg-Pro-Arg-OH), 6 nCi/measurement point 33P-gamma ATP, 0.008% NP40, 1.5% (v/v) dimethylsulfoxide]. The reaction was stopped by the addition of 20 μl of a suspension of streptavidine coated PVT-SPA-beads (0.15 mg/measurement point, from Amersham Biotech) in an aqueous
EDTA/ATP-solution (20 mM EDTA, 50 μM ATP, 1 % (v/v) Triton X-100 in PBS).
The resulting mixture was incubated further 16 h at 22°C to allow the binding of the biotinylated peptide to the streptavidine coated PVT-SPA-beads and to allow the sedimentation of the beads. Subsequently the amount of 33P incorporated into the substrate peptide was evaluated by scintillation measurement in a Topcount NXT (Perkin-Elmer). Chk2 kinase assay
Recombinant Chk2-His6-fusion protein, expressed in insect cells (Sf-9) and purified by Ni-NTA affinity chromatography was used as kinase. Alternatively, commercially available GST-Chk2-fusion protein (Upstate Biotechnology, Dundee, Scotland) can be used. As substrate for the kinase reaction the biotinylated peptide biotin-Arg-Ser-Gly-Leu-Tyr-Arg-Ser-Pro-Ser-Met-Pro-Glu-Asn-Leu-Asn Arg-Pro-Arg-OH was used which can be purchased e.g. from the company Biosyntan GmbH (Berlin-Buch, Germany).
Chk2 (400 ng/measurement point) was incubated for 60 min at 22DC in the presence of different concentrations of test compounds (0 μM and concentrations in the range 0.001 - 30 μM) in 30 μl assay buffer [50 mM Hepes/NaOH pH7,5, 10 mM MgCI2, 1 mM MnCI2, 0.1 mM sodium ortho-vanadate, 1.0 mM dithiothreitol, 1.5 μM adenosine-tri-phosphate (ATP), 8 μM substrate peptide (Biotin-Arg-Ser-Gly-Leu-Tyr-Arg-Ser- Pro-Ser-Met-Pro-Glu-Asn-Leu-Asn-Arg-Pro-Arg-OH), 15 nCi/measurement point 33P-gamma ATP, 0.008% NP40, 1.5% (v/v) dimethylsulfoxide]. The reaction was stopped by the addition of 20 μl of a suspension of streptavidine coated
PVT-SPA-beads (0.25 mg/measurement point, from Amersham Biotech) in an aqueous EDTA/ATP-solution (20 mM EDTA, 50 μM ATP, 1 % (v/v) Triton X-100 in PBS).
The resulting mixture was incubated further 16 h at 22°C to allow the binding of the biotinylated peptide to the streptavidine coated PVT-SPA-beads and to allow the sedimentation of the beads. Subsequently the amount of 33P incorporated into the substrate peptide was evaluated by scintillation measurement in a Topcount NXT (Perkin-Elmer). FACS-Assay
Human HeLa (ATCC CCL-2) cervix adenocarcinoma cells were plate out to a density of 3000 cells / cm2 in DMEM medium containing 10% FCS in 6-well plates. After 48 h incubation the medium was exchange for DMEM medium supplemented with 10% FCS and 5 μg/ml bleomycine sulfate. After 18 h incubation the test compounds were added to final concentrations of 0.03 μM, 0.1μM, 0.3 μM, 1μM,3 μM, 10 μM, or 30 μM. After a further incubation of 24 h or 48 h the cells were collected by trypsinisation, permeablelised and fixed in 70 % ethanol . The DNA was stained with propidium iodide and the cellular DNA-content was measured by a Fluorescence Activated Cell Scan (FACS). The portion of cells with a cellular DNA-content corresponding to the G2 and M phases of the cell cycle was evaluated to judge the effect of the test compound on the bleomycine induced G2/M arrest of the cells.
Expression and purification of Chk1 and Chk2
The coding sequences were cloned by RT-PCR and nested PCR from commercially available polyA-RNA. The primers used for this purpose were designed according to the sequence information in Genebank (AF 016582 for Chk1, AF086904 for Chk2). In preparation for the C-terminal His6-fusion in the respective second PCRs 3'-primers were used, which removed the stop codon at the end of the coding sequence of Chk1 and Chk2 by mutation. Additional restriction sites were added to the primers (EcoRI-sites for the 5'-primers and Hindlll-sites for the 3'-primers).
The cDNAs were cloned into the vector pT7-Blue T (Novagen). To introduce the His6-sequence at the C-terminus of Chk1 and Chk2 EcoRI/Hindlll fragments from these pT7-Blue plasmids were cloned into the bacterial expression vector pET23a. From these pET23a-Chk1 und pET23a-Chk1 vectors DNA fragments coding for Chk1-His6 or Chk2-His6 were excised and inserted into the baculovirus-transfer-vector pVL1392. The generated vectors were transfected into Sf-9 cells with AcNPV baculovirus genomic DNA (BaculoGold Transfection Kit, Pharmingen). The viruses produced by this procedure were plaque-purified and amplified for further infections.
Recombinant Chkl-Hisβ-fusion protein and recombinant Chk2-His6-fusion protein were produced in Sf-9-cells. The Sf-9-cells were infected with the viruses at a MOI (Multiplicity of infectivity) = 1 and subsequently cultivated for 3 days in TNM-FH-medium. After lysis of the cells and sedimentation of the cell debris by centrifugation (20000 x g) the fusion proteins were purified from the supernatant by Ni-NTA affinity chromatography (Superflow from QIAGEN, Hilden, Germany) and dialysed into 50 mM Tris/HCI buffer (pH 7.5) containing 150 mM NaCl and 2 mM EDTA. The protein solution was shock frozen and stored at -80DC.
Results
Compounds, which preferentially inhibit Chk activity are shown in figure 2.
An overview of the results of the inhibition IC5o in nM are presented in the table 2 below:
Table 2:
Figure imgf000063_0001
References:
Chaturvedi, P. et al. (1999), Oncogene 18, 4047-4054.
Dasika, G.K: et al. (1999), Oncogene 18, 7883-7899.
Hartwell, L.H. et al. (1989), Science 246, 629-634. Hartwell, L.H. und Kastan, M.B. (1994). Science 266, 1821-1828.
Hirao, A. et al. (2000), Science 287, 1824-1827.
Jackson, J. R. et al. (2000),Cancer Res. 60, 566-572.
Koniaras, K. et al. (2001), Oncogene 20, 7453-7463.
Levine, A.J. (1997), Cell 88, 323-331. Liu, Q. et al. (2000), Genes Dev. 14,1448-1459.
Matsuoka, S. et al. (1998), Science 282, 1893-1897.
O'Connor, P. M., und Fan, S. (1996). Prog. Cell Cycle Res. 2, 165-173.
Sanchez, Y. et al. (1997), Science 277, 1497-1501.
Takai, H. et al. (2000), Genes Dev. 14, 1439-1447.
Inhibition of KDR- kinase activity
KDR kinase assay
Recombinant KDR-GST-fusion protein, expressed in insect cells (Sf-9) and purified by Glutathion affinity chromatography was used as kinase. Alternatively, commercially available GST-KDR-fusion protein (Proqinase, Freiburg i.Brsg., Germany) can be used. As substrate for the kinase reaction the biotinylated copolymer poly-(Glu, Tyr; 4:1) which can be purchased e.g. from the company Cisbiointernational (Marcoule, France).
In a black low volume 384well microtiterplate (Greiner, Frickenhausen, Germany) KDR (enzyme amount depending on lot, adjusted to give an dF of about 300 - 400) was incubated for 20 min at 22°C in the presence of different concentrations of test compounds (0 μM and concentrations in the range 0.001 - 30 μM) in 15 μl assay buffer [50 mM Hepes/NaOH pH7.0, 25 mM MgCI2, 5 mM MnCI2, 0.5 mM sodium ortho-vanadate, 1.0 mM dithiothreitol, 1 μM adenosine-tri-phosphate (ATP), 23.5 μg/ml substrate [biotinylated poly-(Glu, Tyr; 4:1)], 1.5% (v/v) dimethylsulfoxide]. The reaction was stopped by the addition of 5 μl of a solution of the detection reagents [0.3 μg/ml Eu-W1024-labeled antiphosphotyrosine antibody (PT66) (Perkin-Elmer) and 4.125 μg/ml SA-XL- 665 (Cisbiointernational, Marcoule, France)] in an aqueous EDTA -solution (250 mM EDTA, 0.1 % (w/v) bovine serum albumine in 100 mM HEPES/NaOH pH 7.0).
The resulting mixture was incubated further 2 h at 22°C to allow the binding of the biotinylated substrate and product to the SA-XL-665 and the EU labeled anti- phosphotyrosine antibody. Subsequently the amount of phosphate incorporated into the substrate was evaluated by resonance energy transfer measurement in a HTRF reader (Discovery, Perkin-Elmer).
The IC50 values are determined from the inhibitor concentration that is necessary to inhibit the phosphate incorporation to 50% of the uninhibited incorporation after removal of the blank reading (EDTA-stopped reaction).
Results:
Compounds, which preferentially inhibit Akt and/or Pdk and the VEGF-R activity are shown in figure 3.
An overview of the results of the inhibition IC50 in nM are presented in the table 3 below:
Table 3:
Figure imgf000065_0001
Figure imgf000066_0001
Further, the invention is explained in more detail by the enclosed drawings and examples.
Figures:
Figure 1: preferred compounds inhibiting preferentially Akt, Pdk kinases
Figure 2: preferred compounds inhibiting preferentially Chk kinases
Figure 3: preferred compounds inhibiting preferentially Akt and/or Pdk and VEGF-
R kinases
The following examples demonstrate the feasability of the disclosed invention, without restricting the inventon to these disclosed examples.
Synthetic Schemes
Scheme 1:
Figure imgf000067_0001
Scheme 2:
Figure imgf000067_0002
X = 0-6 Scheme 3:
Figure imgf000068_0001
Scheme 4:
Figure imgf000068_0002
4-AKT
Where R' = Ci-eAlkyl and PG = -NHCOOR6
Scheme 4A
Figure imgf000068_0003
Where R' = d-6Alkyl Scheme 4B
Figure imgf000069_0001
Where R' = Ci-eAlkyl
Scheme 4C
Figure imgf000069_0002
Where R' = Ci-eAlkyl
Scheme 4D
Figure imgf000069_0003
Where R' = Cι-6Alkyl Scheme 4E
Figure imgf000070_0001
Where R' = d-6Alkyl
Scheme 4F
Figure imgf000070_0002
Where R" = Ci-eAlkyl and PG = -NHCOOR6
Scheme 5
Figure imgf000070_0003
6-AKT
Where R8 and R9 are as described in the claims.
Scheme 6
Figure imgf000070_0004
Where R is as described in the claims.
Scheme 7 u R' O
H ' fl 1 6 N HCI, acetone
✓-\ ,O e THF, RT, o/n VN λO e ^
N=C-=0 - O R' O 02N 02N 10-AKT 2 H2, Pd/C, MeOH 9-AKT
Figure imgf000071_0001
Where R' is hydrogen or methyl.
Scheme 8
Figure imgf000071_0002
Where R5 is as described in the claims and PG = -NHCOOR6
Scheme 9
Figure imgf000071_0003
Where R' is d-6Aklylaryl or d-eAlkylheteroaryl. Scheme 10
CsC03, DMF, heat R8
PG. PG.
N— R'- R" + HNR8R9 N— R'- N H H
R9
19-AKT 20-AKT
Where R' is Ci-e lkyl, R" is halogen, R8 and R9 are as described in the claims and PG = -NHCOOR6 Scheme 11
Figure imgf000072_0001
21 -AKT
HCI, H20, MeCN, reflux
Figure imgf000072_0002
Where R' is Ci-eAlkyl; A, B, R8, R9 are as described in the claims and PG = R6 as described in the claims.
Scheme 12
Figure imgf000072_0003
25-AKT 26-AKT
Where R' is Ci-eAlkyl; and R1, A and B are as described in the claims. Scheme 13
Figure imgf000073_0001
26'-AKT 27-AKT
Where R' is Ci-eAlkyl and R" is cycloalkyl ring, heteroaryl or aryl; and R1, A and B are as described in the claims.
Scheme 14
Figure imgf000073_0002
Where R1 and A are as described in the claims.
Scheme 15
Figure imgf000073_0003
Where R" is Ci-βAlkyl and R and R5 are as described in the claims. Examples
A. Synthesis of Compounds
The following Examples have been synthesized according to the above mentioned schemes.
A1
5-Bromo-4-(2-(1H-imidazol-4-yI)-ethylamino)-2-(4-pyrrolidin-1-ylmethyl- phenylamino)-pyrimidine
1a) 5-Bromo-2,4-dichloropyrimidine
To 5-bromouracil (50 g) were sequentially added Λ/,Λ/-diethylaniline (60 mL) and phosphoryl chloride (120 mL), and the mixture was refluxed for 5 h. The volatiles were removed by distillation, the residue poured into ice water and the mixture extracted with methyl fetτ-butyl ether. The combined extracts were washed with brine, dried (Na2SO ) and filtered through Celite. Distillation of the crude product gave the title compound (63.4 g).
1H NMR (300 MHz, CDCI3): δ/ppm = 8.69 (s, 1H).
1b) 5-Bromo-4-(2-(7H-imidazol-4-yl)-ethylamino)-2-chloro-pyrimidine
To a solution of 5-bromo-2,4-dichloropyrimidine (4.56 g) and triethylamine (3 mL) in acetonitrile (20 mL) 2-(7/-/-imidazol-4-yl)-ethylamine (2.45 g) was added portionwise at 0 °C, and the suspension stirred at room temperature overnight. The reaction mixture was partitioned between ethyl acetate and brine, the aqueous phase extracted with additional ethyl acetate, the combined organic phases dried (Na2SO4) and evaporated, which gave, after chromato- graphy on silica using dichloromethane/methanol, the title compound (4.41 g).
1H NMR (300 MHz, CD3OD): δ/ppm = 2.91 (t, 2H, J=7 Hz), 3.73 (t, 2H, J=7 Hz), 6.87 (s, 1H), 7.61 (s, 1H), 8.11 (s, 1H). 1 c) 4-Pyrrolidin-1 -ylmethyl-phenylamine
To a suspension of sodium hydride (60% in oil, 220 mg) in THF (5 mL) pyrrolidine (391 mg) was added, the mixture stirred at r.t. for 6 h, a solution of 1- bromomethyl-4-nitro-benzene (1.08 g) in THF (8 mL) added and stirred overnight. The reaction was quenched with water and extracted with ethyl acetate, the organic phase dried (Na2SO4) and evaporated, which gave, after chromatography on silica using dichloromethane/methanol, 1-(4-nitro-benzyl)- pyrrolidine (690 mg).
1H NMR (300 MHz, CDCI3): δ/ppm = 1.84 (m, 4H), 2.58 (m, 4H), 3.77 (s, 2H), 7.61 (dbr, 2H, J=9 Hz), 8.22 (dbr, 2H, J=9 Hz).
To a solution of 1-(4-nitro-benzyl)-pyrrolidine (1.37 g) in ethanol (66 mL) tin(ll)- chloride dihydrate (9.0 g) was added portionwise and the resulting mixture refluxed for 2 h. The reaction mixture was partitioned between ethyl acetate and saturated aqueous sodium bicarbonate solution, the aqueous phase extracted with additional ethyl acetate, the combined organic phases dried (Na2SO4) and evaporated, which gave, after chromatography on silica using dichloromethane/methanol, the title compound (432 mg).
1H NMR (300 MHz, CD3OD): δ/ppm = 1.85 (m, 4H), 2.65 (m, 4H), 3.61 (s, 2H), 6.72 (d, 2H, J=9 Hz), 7.11 (d, 2H, J=9 Hz).
1d) 5-Bromo-4-(2-(7H-imidazol-4-yl)-ethylamino)-2-(4-pyrrolidln-1-ylmethyl- phenylamino)-pyrimidine
A mixture of 5-bromo-4-(2-( /-/-imidazol-4-yl)-ethylamino)-2-chloro-pyrimidine (60 mg), 4-pyrrolidin-1 -ylmethyl-phenylamine (35 mg) and hydrochloric acid (37% in water, 40 μL) in methanol (2 mL) was refluxed overnight. The reaction mixture was partitioned between ethyl acetate and saturated aqueous sodium bicarbonate solution, the organic phase dried (Na2SO4) and evaporated, which gave, after chromatography on silica using dichloromethane/methanol, the title compound (4 mg). 1H NMR (400 MHz, CD3OD): δ/ppm = 2.09 (m, 4H), 3.02 (t, 2H, J=7 Hz), 3.31 (m, 4H), 3.79 (t, 2H, J=7 Hz), 4.30 (s, 2H), 7.11 (s, 1H), 7.40 (d, 2H, J=9 Hz), 7.76 (d, 2H, J=9 Hz), 7.97 (s, 1H), 8.19 (s, 1 H).
A2
2-(4-(Aminomethyl)-phenylamino)-4-(prop-2-ynylam»no)-5-trifluoromethyl- pyrimidine
2a) 2,4-Dichloro-5-trifluoromethyl-pyrimidine
To 5-trifluoromethyluracil (25 g) were sequentially added Λ/,Λ/-diethylaniline (25 g) and phosphoryl chloride (94 g), and the mixture was refluxed for 18 h. After cooling to r.t. the solution was poured onto ice (100 g), stirred for 10 min. and extracted with diethyl ether. The combined organic phases were washed with saturated aqueous sodium carbonate solution and water, dried (Na2SO4), and filtered. After removal of most of the ether, distillation of the residue at 190 °C and 860 to 300 mbar gave the title compound (5.8 g).
1H NMR (300 MHz, CDCI3): δ/ppm = 8.83 (s, 1H).
2b) 2-Chloro-4-(prop-2-ynylamlno)-5-trifluoromethyl-pyrimldine
To a solution of 2,4-dichloro-5-trifluoromethyl-pyrimidine (3.47 g) in acetonitrile (16 mL) a solution of propargylamine (1.76 g) in acetonitrile (16 mL) was added dropwise at 0 °C, the mixture warmed to r.t. and stirred at r.t. for 48 h. The suspension was diluted with ethyl acetate, washed with brine, dried (Na2SO ), and evaporated. Purification by flash chromatography on silica using hexane/methyl terf-butyl ether gave the title compound (1.97 g).
1H NMR (400 MHz, CDCI3): δ/ppm = 2.34 (t, 1H, J=1.5 Hz), 4.37 (dd, 2H, J=1.5/5 Hz), 5.53 (brs, 1 H), 8.33 (s, 1 H). 2c) 2-(4-(Aminomethyl)-phenylamino)-4-(prop-2-ynylamino)-5- trifluoromethyl-pyrimidine
A mixture of 2-chloro-4-(prop-2-ynylamino)-5-trifluoromethyl-pyrimidine (235 mg), Λ/-(4-aminobenzyl)-2,2,2-trifluoro-acetamide (410 mg) and hydrochloric acid (37% in water, 0.2 mL) in methanol (5 mL) was refluxed for 1 h. The reaction mixture was partitioned between ethyl acetate and saturated aqueous sodium bicarbonate solution, the aqueous phase extracted with ethyl acetate, the combined organic phases dried (Na2SO4), concentrated, filtered through silica using dichloromethane/methanol, and the filtrate evaporated. To a solution of the residue in methanol (9 mL), tetrahydrofuran (9 mL) and diethyl ether (4.5 mL) was added lithium hydroxide (150 mg) and the mixture was refluxed for 6 h, after which it was partitioned between ethyl acetate and saturated aqueous sodium bicarbonate solution. The aqueous phase was extracted with additional ethyl acetate, the combined organic phases dried (Na2SO ) and evaporated, which gave, after chromatography on silica using dichloromethane/methanol, the title compound (120 mg).
1H NMR (300 MHz, CD3OD): δ/ppm = 2.55 (t, 1H, J=1.5 Hz), 4.07 (s, 2H), 4.26 (d, 2H, J=1.5 Hz), 7.39 (d, 2H, J=8 Hz), 7.86 (d, 2H, J=8 Hz).
A3
/V-(3-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4- pyrimidinyl)amino)propyl)-7 /-pyrrole-2-carboxamide
3a) (3-((5-bromo-2-chloro-4-pyrimidinyl)amino)propyl)-carbamic acid tert- butyl ester
To a solution of 5-bromo-2,4-dichloro-pyrimidine (1.4 g) in acetonitrile (10 mL) at 0 °C was added triethylamine (0.94 mL) and 3-aminopropylcarbamic acid-1 ,1- dimethylethyl ester (1.0 g). After removing the cooling bath the reaction mixture was stirred overnight at room temperature. The reaction mixture was concentrated and to the residue water (20 mL) was added. The precipitate was collected, washed with water and ether to afford the title compound (1.8 g). 1H NMR (400 MHz, DMSO-d6): δ/ppm = 1.34 (s, 9H), 1.62 (m, 2H), 2.93 (m, 2H), 3.36 (m, 2H), 6.78 (t, 1 H), 7.64 (t, 1H), 8.22 (s, 1 H).
3b) 4-((4-((3-aminopropyl)amino)-5-bromo-2-pyrimidinyl)amino)- benzenesulfonamide hydrochloride
To a solution of 4-aminobenzenesulfonamide (190 mg) in acetonitrile (20 mL), hydrochloric acid solution (4M in dioxane, 0.3 mL) and water (0.3 mL) was added (3-((5-bromo-2-chloro-4-pyrimidinyl)amino)propyl)-carbamic acid-1 ,1-dimethylethyl ester (360 mg). The resulting mixture was refluxed overnight. The precipitate was collected and washed with acetonitrile and methanol to afford the title compound (320 mg).
1H NMR (400 MHz, DMSO-d6): δ/ppm = 1.86 (m, 2H), 2.78 (m, 2H), 3.51 (m, 2H), 7.23 (s, 2H), 7.75 (d, 2H), 7.79 (d, 2H), 7.96 (m, 3H), 8.19 (s, 1 H), 10.38 (t, 1H).
3c) N-(3-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4- pyrimidinyl)amino)propyl)-7H-pyrrole-2-carboxamide trifluoroacetate 4-((4-((3-aminopropyl)amino)-5-bromo-2-pyrimidinyl)amino)-benzenesulfonamide (120 mg) was suspended in dimethylformamide (5 mL). 2-Pyrrolecarboxylic acid (50 mg), O-(7-azabenzotriazol-1-yl)-/V,Λ/,Λ/',Λ/'-tetramethyluronium hexafluorophosphate (180 mg), and diisopropylethylamine (0.3 mL) were added and the resulting mixture was stirred at room temperature for 15 min. Purification by HPLC chromatography using acetonitrile/water gave the title compound (65 mg).
1H NMR (400 MHz, DMSO-d6): δ/ppm = 1.78 (m, 2H), 3.27 (m, 2H), 3.44 (m, 2H), 6.03 (d, 1H), 6.71 (s, 1 H), 6.80 (s, 1 H), 7.14 (s, 2H), 7.42 (t, 1H), 7.68 (d, 2H), 7.83 (d, 2H), 8.04 (t, 1 H), 8.11 (s, 1 H), 9.78 (s, 1 H), 11.39 (s, 1 H). A4
Λ/-[3-[[(2/?)-2-Amino-1-oxo-3-phenylpropyl]amino]-5-[[5-bromo-4-(prop-2- ynyloxy)pyrimidin-2-yl]amino]phenyl]pyrrolidine-1-carboxamide
4a) Methyl3-amino-5-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin 2yl]amino]benzoate
A mixture of 5-bromo-2-chloro-4-(prop-2-ynyloxy)pyrimidine (15 g), methyl 3,5- diaminobenzoate (45 g) and concentrated hydrochloric acid (15 ml) in methanol (600 ml) was stirred at 65°C for 8 h. After concentration to half the volume water was added and the precipitate collected by filtration. The precipitate then was treated with sodium hydroxide solution (1 n) and dichloromethane. The organic phase then was washed with water and brine, dried (Na2SO ) and evaporated to dryness to give the title compound (13.8 g). Mp.: 207.5-209 °C
4b) Methyl 5-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-3-[[(2R)-2-
[[(1,1-dimethylethoxy)carbonyl]amino]-1-oxo-3- phenylpropyl]amino]benzoate
Λ/-BOC-D-phenylalanine (3.3 g), 1-hydroxy-1/-/-benzotriazole hydrate(1.9 g) and /V-[3-(dimethylamino)propyl]-/V-ethylcarbodiimid hydrochloride (2.37 g) were stirred in DMF (30 ml) for 30 minutes. Then methyl 3-amino-5-[[5-bromo-4- (prop-2-ynyloxy)pyrimidin-2-yl]amino]benzoate (3.88 g) were added and the mixture stirred over night. Then ethyl acetate (500 ml) was added and the reaction mixture washed subsequently with hydrochloric acid (0.1 n), saturated NaHCO3-solution, water and brine. After drying (Na2SO4) the organic phase was evaporated and the residue subjected to column chromatography (ethyl acetate/dichloromethane) to yield 5.36 g of the title compound. ESI-MS: 624 and 626 (M+)
4c) 5-[[5-Bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-3-[[(2R)-2-[[(1 ,1 - dimethylethoxy)carbonyl]amino]-1-oxo-3-phenylpropyl]amino]benzoic acid
Methyl 5-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-3-[[(2R)-2-[[(1 ,1- dimethylethoxy)carbonyl]amino]-1 -oxo-3-phenylpropyl]amino]benzoate (1.0 g) was stirred in a mixture of tetrahydrofuran (20 ml), methanol (20 ml)and sodium hydroxide solution (2 n; 20 ml) for 48 h. After evaporation water (50 ml) was added to the residue. On neutralisation with hydrochloric acid (1 n) a precipitate formed. The precipitate was subjected to chromatography on silica gel (hexanes/ethyl acetate/methanol) to yield the title compound (450 mg). ESI-MS: 610 and 612 (M+)
4d) 1 ,1 -Dimethylethoxy [(1 R)-2-[[3-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2- yl]amino]-5-[[(pyrrolidin-1-yl)carbonyl]amino]phenyl]amino]-2-oxo-1- (phenylmethyl)ethyl]carbamate
5-[[5-Bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-3-[[(2f?)-2-[[(1 ,1- dimethylethoxy)carbonyl]amino]-1 -oxo-3-phenylpropyl]amino]benzoic acid (200 mg), diphenylphosphorylazide (0.75 ml) and triethylamine (0.67 ml) were refluxed in toluene (40 ml) for 1.5 h. Then pyrrolidine (0.26 ml) was added and the mixture refluxed for additional 2 h. After cooling the reaction mixture was diluted with ethyl acetate (50 ml) and subsequently washed with saturated NaHCO3-solution, water and brine. After drying (Na2SO4) and evaporation the residue was subjected to chromatography on silica gel (hexanes/ethyl acetate) to yield the title compound (126 mg). ESI-MS: 678 and 680 (M+)
4e) Λ/-[3-[[(2R)-2-Amino-1-oxo-3-phenylpropyl]amino]-5-[[5-bromo-4-(prop-2- ynyloxy)pyrimidin-2-yl]amino]phenyl]pyrrolidine-1-carboxamide
1 ,1 -Dimethylethoxy [(1f?)-2-[[3-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2yl]amino]- 5-[[(pyrrolidin-1 -yl)carbonyl]amino]phenyl]amino]-2-oxo-1 (phenylmethyl)ethyl] carbamate (105 mg) and sulfuric acid (0.5 ml; 2 n) were stirred in dioxane (5 ml) at 85°C for 3.5 h. After cooling and dilution with water saturated NaHCO3- solution was added and the resulting precipitate collected by filtration yielding the title compound (76 mg). ESI-MS: 578 and 580 (M+) A4A
Synthesis of [3-[[5-bromo-2-[[3-[(1-pyrrolidinylcarbonyl) amino]phenyl]amino]-4-pyrimidinyl]amino]propyl]-carbamic acid ethyl ester
To a solution of Λ/-(3-((4-((3-aminopropyl)amino)-5-bromo-2-pyrimidinyl)amino) phenyl)-1 -pyrrolidinecarboxamide (150 mg, 0.30 mmol) in pyridine (5mL) was added ethyl chloroformate (38.5 mg, 0.35 mmol) at 0°C under N2. The resulting reaction mixture was stirred at 0°C for 1h and then was stirred at room temperature overnight. The mixture was washed with water (3 x 50 mL). Then the reaction mixture was concentrated. Purification by HPLC chromatography using acetonitrile/water gave the title compound (10 mg).
1H NMR (400 MHz, DMSO-d6): δ/ppm = 0.79(t, 3H), 1.38 (t, 2H), 1.48 (m, 4H), 2.65 (m, 2H), 3.00 (m, 4H), 3.19 (m, 2H), 3.59 (m, 2H), 6.78 (m, 1 H), 6.85 (m, 2H), 7.57 (s, 1 H), 7.82 (m, 2H), 8.23 (m, 1 H), 10.08 (s, 1H)
A4B
Synthesis of N-[3-[[5-bromo-4-[[3-[(propylsulfonyl)amino]propyl]amino]-2- pyrimidinyl]amino]phenyl]-1-pyrrolidinecarboxamide
To a solution of Λ/-(3-((4-((3-aminopropyl)amino)-5-bromo-2-pyrimidinyl)amino) phenyl)-1 -pyrrolidinecarboxamide (150 mg, 0.30 mmol) in dichloromethane (4mL) was added DIEA (0.16 mL, 0.92 mmol) and DMAP (1.4 mg, 0.011 mmol) at 0°C, then a solution of 1-propanesulfonyl chloride (51 mg, 0.36 mmol) in dichloromethane (5mL) was added. The resulting reaction mixture was stirred at 0°C for 1h and at room temperature overnight. The reaction mixture was concentrated. Purification by HPLC using acetonitrile/water gave the title compound (67mg).
1H NMR (400 MHz, DMSO): δ/ppm = 0.82 (t, 3H), 1.61 (m, 2H), 1.76 (m, 2H), 1.79 (m, 4H), 2.80 (m, 2H), 2.90 (m, 2H), 3.31 (m, 4H), 3.51 (m, 2H), 7.09 (m, 1H), 7.18 (m, 2H), 7.89 (s, 1H), 8.11 (s, 2H), 8.50 (m, 1H), 10.31 (s, 1H) A4C
Synthesis of N-[3-[[5-bromo-4-[[3-[[(phenylamino)carbonyl]amino] propyl]amino]-2-pyrimidinyl]amino]phenyl]-1-pyrrolidinecarboxamide
To a suspension of Λ/-(3-((4-((3-aminopropyl)amino)-5-bromo-2-pyrimidinyl) amino)phenyl)-1-pyrrolidinecarboxamide (100 mg, 0.2 mmol) and DIEA (0.14mL, O.δmmol) in 1,4-dioxane (5mL) was added phenyl isocyanate (35 mg, 0.3mmol). The resulting solution was stirred overnight and concentrated. The crude residue was directly purified by prep HPLC using acetonitrile/water to give the title compound (68 mg).
1H NMR (400 MHz, DMSO-d6), δ/ppm = 1.71 (m, 2H), 1.84 (m, 4H), 3.09 (m, 2H), 3.36 (m, 4H), 3.48 (m, 2H), 6.21 (t, 1 H), 6.83 (t, 1 H), 7.05 (m, 1H), 7.19 (m, 4H), 7.36 (m, 2H), 7.84 (br s, 1 H), 7.92 (s, 1 H), 8.16 (s, 2H), 8.47 (s, 1 H), 9.71 (s, 1 H).
A4D
Synthesis of N-[3-[[5-bromo-4-[[3-[[(ethylamino)thioxomethyl]amino] propyl]amino]-2-pyrimidinyl]amino]phenyl]-1 -pyrrolidinecarboxamide
A solution of Λ/-(3-((4-((3-aminopropyl)amino)-5-bromo-2-pyrimidinyl) amino)phenyl)-1 -pyrrolidinecarboxamide (100 mg, 0.20 mmol) and DMF (5 mL) was treated with DIEA (0.1 mL, 0.6 mmol, 3eq) and ethylthioisocyanate (15 mg, 0.17 mmol, 0.9 eq). The resulting mixture was stirred at RT for 2hr. Then the crude mixture was purified by HPLC using acetonitrile/water to afford the title compound (82 mg).
1H NMR (400 MHz, DMSO-d6): δ/ppm = 1.02 (t, 3H), 1.74 (m, 2H), 1.82 (m, 4H), 3.30-3.48 (m, 8H), 7.04-7.16 (m, 3H), 7.37 (m, 2H), 7.88 (s, 1 H), 8.08 (m, 2H). A4E
Synthesis of [3-[[5-bromo-2-[[3-[(1-pyrrolidinylcarbonyl)amino]phenyl] amino]-4-pyrimidinyl]amino]propyl]-carbamothioic acid S-ethyl ester
A solution of Λ/-(3-((4-((3-aminopropyl)amino)-5-bromo-2-pyrimidinyl)amino) phenyl)-1 -pyrrolidinecarboxamide (150 mg, 0.30 mmol), DMF (1.5 mL) and dichloromethane (5 mL) was treated with DIEA (0.2 mL, 1.15 mmol, 4 eq.) and the was treated dropwise with a solution of ethyl chlorothioformate (41 mg, 0.33 mmol, 1.1 eq) and dichloromethane (1 mL). The resulting mixture was stirred at rt. for 30 mins. Then the reaction mixture was diluted with dichloromethane (30 mL),washed with water (3 x 20 mL) and concentrated. The crude product was purified by chromatography on SiO2 using ethyl acetate/methanol to afford the titile compound (112 mg).
1H NMR (400 MHz, DMSO-d6): δ/ppm = 1.14 (t, 3H), 1.68 (m, 2H), 1.82 (m, 4H), 2.74 (q, 2H), 3.13 (m, 2H), 3.35 (m, 4H), 3.42 (m, 2H), 6.89 (t, 1 H), 6.94 (d, 1 H), 7.05 (t, 1H), 7.23 (d, 2H), 7.86 (s, 1 H), 7.95 (m, 2H), 8.12 (t, 1 H), 9.06 (s, 1H).
A4F
Synthesis of N-[3-[[4-[[3-[(aminosulfonyl)amino]propyl]amino]-5-bromo-2- pyrimidinyl]amino]phenyl]-1 -pyrrolidinecarboxamide
Chloro[[(1 ,1-dimethylethoxy)carbonyl]amino]-sulfane dioxide was prepared by adding chlorosulfonyl isocyanate (32 mg, 0.23 mmol, 1.0 eq.) to a cooled solution of fetf-butyl alcohol (17 mg, 0.23 mmol, 1.0eq.) and dichloromethane (2 mL) in an ice-water bath. The resulting mixture was stirred at 0-5°C for 2-3hr. The solution was then treated with a solution of Λ/-(3-((4-((3- aminopropyl)amino)-5-bromo-2-pyrimidinyl)amino)phenyl)-1- pyrrolidinecarboxamide (100 mg, 0.20 mmol, 1eq.) and dichloromethane (5 mL). DMAP (20 mg, 0.16 mmol) was then added followed by the dropwise addition of DIEA (0.1 mL, 0.57 mmol). The mixture was stirred at RT for overnight. The reaction mixture was concentrated in vacuo. The residue was dissolved in TFA (2 mL), and purified by HPLC using acetonitrile/water to afford the title compound (30 mg).
1H NMR (400 MHz, DMSO-d6): δ/ppm = 1.76 (m, 2H), 1.82 (m, 4H), 2.92 (m, 2H), 3.36 (m, 4H), 3.45 (m, 2H), 6.48 (s, 2H), 7.04 (d, 1 H), 7.14 (t, 1 H), 7.21 (d, 2H), 7.82 (s, 1 H), 8.05 (m, 2H).
A 5 Λ/-(3-aminophenyl)-urea (A5)
Ammonia was bubbled into a solution of 3-nitrophenylisocyanate (1.5 g, 9.1 mmol) for ten minutes. The reaction mixture was then concentrated and the resulting yellow solid was washed with ether (200 mL) to afford Λ/-(3- nitrophenyl)-urea (1.35 g, 7.5 mmol). A solution of Λ/-(3-nitrophenyl)-urea (1.0 g, 5.5 mmol) and methanol (40 mL) was treated with 10% Pd/C (250 mg) and placed under H2 (45 psi) for 2 h. The mixture was then filtered through celite and concentrated to afford Λ/-(3- aminophenyl)-urea (828 mg, 5.5 mmol).
1H NMR (400 MHz, DMSO): δ/ppm = 4.90 (s, 2H), 5.66 (s, 2H), 6.08 (dm, J = 8 Hz, 1 H), 6.43 (dm, J = 8 Hz, 1 H), 6.70 (t, J = 1.6 Hz, 1 H), 6.80 (t, J = 8 Hz, 1 H), 8.13 (s, 1 H).
A 6 (3-aminophenyl)-2-(4-morpholinyl)-carbamic acid ethyl ester
6a) 2-(4-morpholinyl)-(3-nitrophenyl)-carbamic acid ethyl ester
A solution of 3-nitrophenyl isocyanate (0.5 g, 3.0 mmol) and 4-(2- aminoethyl)morpholine (0.5 mL, 3.8 mmol, 1.3 equiv.) in tetrahydrofuran (20mL) was stirred for 3 h. The reaction mixture was concentrated and purified by chromatography (SiO2) using hexane/ethyl acetate to afford 2-(4-morpholinyl)- (3-nitrophenyl)-carbamic acid ethyl ester (0.5 g). 1H NMR (400 MHz, CDCI3): δ/ppm = 2.52 (m, 4H), 2.58 (m, 2H), 3.39 (m, 2H), 3.76 (m, 4H), 5.35 (br s, 1 H), 7.43 (t, 1 H), 7.87 (m, 2H), 8.20 (m, 1H)
6b) (3-aminophenyl)-2-(4-morpholinyl)-carbamic acid ethyl ester A solution of 2-(4-morpholinyl)-(3-nitrophenyl)-carbamic acid ethyl ester (0.5 g, 1.7 mmol) and methanol (50 mL) was treated with 10% Pd/C (150 mg) and placed under H2 (50 psi) for 2 h. The mixture was then filtered through celite and concentrated to afford the title compound (320 mg).
1H NMR (400 MHz, CDCI3): δ/ppm = 2.52 (m, 4H), 2.68 (m, 2H), 3.52 (br s, 2H), 3.74 (m, 4H), 4.31 (m, 2H), 6.39 (m, 1 H), 6.58 (m, 1H), 6.68 (br s, 1 H), 6.94 (br s, 1H), 7.09 (m, 1H).
A 7 3-(3-Aminophenyl)-2,4-imidazolidinedione
7a) [[(3-nitrophenyl)amino]carbonyl]aminoacetic acid methyl ester
To a suspension of 3-nitrophenyl isocyanate (10 g, 61 mmol) and glycine methyl ester hydrochloride (8.4 g, 67 mmol, 1.1 equiv.) in dichloromethane (250 mL) was added triethylamine (10 mL, 72 mmol, 1.2 equiv.) at 0 °C. The resulting solution was stirred at room temperature overnight. The resulting dark brown solution was concentrated and triturated in water to give a light yellow suspension. The suspension was filtered and the filter cake was washed with water and air-dried to give [[[(3-nitrophenyl)amino]carbonyl]aminoacetic acid methyl ester (15 g) in quantitative yield.
1H NMR (400 MHz, DMSO-d6): δ/ppm = 3.64 (s, 3H), 3.89 (d, 2H), 6.67 (t, 1H), 7.52 (t, 1 H), 7.68 (dd, 1 H), 7.76 (dd, 1 H), 8.51 (s, 1 H), 9.38 (br s, 1 H). 7b) 3-(3-Nitrophenyl)-2,4-imidazolidinedione
A suspension of [[[(3-nitrophenyl)amino]carbonyl]aminoacetic acid methyl ester (6.9 g, 27 mmol) in 6/V aqueous hydrochloride solution (40 mL) and acetone (20 mL) was stirred at reflux overnight. The resulting solution was cooled and concentrated. The resulting yellowish suspension was filtered and the filter cake was washed with water (50 mL), aqueous sodium bicarbonate solution (50 mL), and air-dried to afford the title compound (4.4 g).
1H NMR (400 MHz, DMSO-d6): δ/ppm = 4.09 (s, 2H), 7.78 (t, 1 H), 7.89 (dd, 1 H), 8.23 (dd, 1H), 8.31 (d, 1H), 8.49 (br s, 1H).
7c) 3-(3-Aminophenyl)-2,4-imidazolidinedione
A solution of 3-(3-nitrophenyl)-2,4-imidazolidinedione (4.4 g, 20 mmol) and methanol (100 mL) was treated with 10% Pd/C (1.0 g) and placed under H2 (40 psi) for 2 h. The mixture was then filtered through celite and concentrated to afford the title compound (3.8 g).
1H NMR (400 MHz, DMSO-d6): δ/ppm = 4.02 (s, 2H), 5.23 (br s, 2H), 6.39 (d, 1 H), 6.47 (s, 1H), 6.54 (d, 1 H), 7.06 (t, 1 H), 8.19 (br s, 1 H).
A8
D-[2-[(3-Aminophenyl)amino]-2-oxo-1-(phenylmethyl)ethyl]-carbamic acid terf-butyl ester
A solution of 1 ,3-phenylenediamine (1.0 g, 10 mmol, 2 equiv.) and N-tert- butoxycarbonyl-D-phenylalanine hydroxysuccinimide ester (1.8 g, 5 mmol, 1 equiv.) in acetonitrile (40 mL) was stirred overnight. The reaction mixture was concentrated and purified by chromatography (SiO2) using dichloromethane/methanol to afford the title compound (1.2 g).
1H NMR (400 MHz, CDCI3): δ/ppm = 1.43 (s, 9H), 3.14 (m, 2H), 3.71 (br s, 2H), 4.48 (br s, 1H), 5.21 (br s, 1 H), 6.43 (m, 1H), 6.53 (br s, 1H), 7.04 (m, 2H), 7.29 (m, 5H), 7.74 (br s, 1 H). A9 5-bromo-2-chloro-iV-[2-(4-thiazolyl)ethyl]-4-pyrimidinamine
Lithium Aluminum hydride (95%) (1.1 g, 27.5 mmol) was suspended in dry THF (20 mL) and cooled with an ice-water bath. A solution of 1 ,3-thiazol-4-acetonitrile (1.0 g, 8.06 mmol) in THF (10 mL) was added dropwise. The resulting mixture was stirred at room temperature overnight. To the reaction mixture was added water (1 mL), 15% NaOH (1 mL) followed by water (3 mL). The precipitate inorganic solid was filtered, then washed with ethyl acetate (100 mL). The combined organic phase was dried (Na2SO ) and concentrate in vacuo to afford 4-thiazoleethanamine as a brown oil (400 mg, 3.12 mmol). The oil (400 mg, 3.12 mmol) was dissolved in CH3CN (10 mL), treated with Et3N (0.7 mL, 97.5 mmol) and cooled with an ice-water bath. 5-Bromo-2,4-dichloropyrimidine (800 mg, 3.51 mmol) was then added. The resulting mixture was stirred at room temperature overnight. The mixture was dried in vacuo, then purified by chromatograpy (SiO2) using hexane/ethyl acetate to afford the titled compound (110 mg)
1H NMR (400 MHz, CDCI3): δ/ppm = 3.13 (t, 2H), 3.86 (m, 2H), 6.74(t, 1H), 7.11(s, 1 H), 8.12(s, 1 H), 8.83(s, 1 H)
A10
[3-(2-thiazolylamino)propyl]-carbamic acid 1,1-dimethylethyl ester
To a solution of (3-bromopropyl)-carbamic acid 1,1-dimethylethyl ester (1.2 g, 5.0 mmol) and 2-aminothiazole (1.0 g, 10 mmol, 2 equiv.) in DMF 20 (mL) was added Cs2CO3 (2.5 g, 7.7 mmol, 1.5 equiv.). The resulting mixture was heated at 85 °C under N2 overnight. The reaction mixture was diluted with ethyl acetate (200 mL), washed with water (3 x 200 mL), and brine (200 mL). The organic phase was dried over Na2SO4, then concentrated in vacuo to afford an oil. The crude product was purified by chromatography (SiO2) using hexane/ethyl acetate to afford the title compound as a light yellow solid (300 mg). 1H NMR (400 MHz, DMSO-d6): δ/ppm = 1.37 (s, 9H), 1.65 (m, 2H), 2.95 (m, 2H), 3.14 (m, 2H), 6.57 (d, 1 H), 6.83 (t, 1 H), 6.98 (d, 1 H), 7.46 (t, 1 H)
A11 N-[3-[[5-bromo-4-[[3-oxo-3-(propylamino)propyl]amino]-2- pyrimidinyl]amino]phenyl]-1 -pyrrolidinecarboxamide
11a)Λ/-[5-bromo-2-[[3-[(1-pyrrolidinylcarbonyl)amino]phenyl]amino]-4- pyrimidinyl]-β-alanine To a solution of 5-bromo-2,4-dichloropyrimidine (1.0 g, 4.4 mmol, 1 equiv.) in acetonitrile (10 mL) at 0°C was added triethylamine (0.672 mL, 4.8 mmol, 1.1 equiv.) and H-beta-Ala-OtBu HCI (0.8 g, 4.4 mmol, 1 equiv.). After removing the cooling bath the reaction mixture was stirred at room temperature overnight. The reaction mixture was concentrated and to the residue water (20 mL) was added. The precipitate was collected, washed with water and ether to afford Λ/-(5-bromo- 2-chloro-4-pyrimidinyl)-β-alanine 1 ,1-dimethylethyl ester (0.52 g).
To a solution of Λ/-(5-bromo-2-chloro-4-pyrimidinyl)-D-alanine 1 ,1-dimethylethyl ester (348 mg, 1.2 mmol, 1 equiv.) in acetonitrile (10 mL) was added water (1.0 mL), 4.0M HCI in dioxane (1.0 mL) and Λ/-(3-aminophenyl)-1- pyrrolidinecarboxamide (520 mg, 2.5 mmol, 2.1 equiv.). The resulting mixture was stirred at 80 °C overnight. The white suspension was filtered and washed with acetonitrile to afford the title compound (500 mg).
1H NMR (400 MHz, DMSO): δ/ppm = 2.15 (t, 4H), 2.79 (t, 2H), 3.55 (t, 4H), 3.89 (m, 2H), 7.45 (m, 3H), 8.10 (s, 1 H), 8.40 (d, 2H), 8.80 (t, 1H), 10.65 (s, 1 H) 11b) N-[3-[[5-bromo-4-[[3-oxo-3-(propylamino)propyl]amino]-2-pyrimidinyl] amino]phenyl]-1 -pyrrolidinecarboxamide
To a solution of Λ/-[5-bromo-2-[[3-[(1-pyrrolidinylcarbonyl)amino]phenyl]amino]-4- pyrimidinyl]-π-alanine (200 mg, 0.45 mmol) in DMF (20 mL) was added O-(7-aza- benzotriazol-1-yl)-Λ/,Λ/,Λ/',Λ/'-tetramethyluronium hexafluorophosphate (243 mg, 0.64 mmol, 1.4 equiv.), diisopropylethylamine (0.46 mL, 2.64 mmo, 5.9 equiv.l) and propylamine (32 mg, 0.54 mmol, 1.2 equiv.). The resulting mixture was stirred at room temperature for 20min. Purification by HPLC chromatography using acetonitrile/water gave the title compound (40mg).
1H NMR (400 MHz, DMSO-d6): δ/ppm = 0.50 (t, 3H), 1.07 (m, 2H), 1.54 (t, 4H), 2.16 (t, 2H), 2.70 (m, 2H), 3.08 (t, 4H), 3.45 (m, 2H), 6.80 (d, 1H), 6.92 (t, 1H), 7.02 (d, 1 H), 7.63 (s, 1 H), 7.69 (t, 1H), 7.91 (s, 1 H), 7.96 (s, 1 H), 8.39 (t, 1 H), 10.13 (s, 1 H)
A12
W-(3-((4-(((3-aminophenyl)methyl)amino)-5-bromo-2-pyrimidinyl) amino)phenyl)-1 -pyrrolidinecarboxamide (ZK 822797/26-AKT) (SY)
Λ/-(3-((5-bromo-4-(((3-nitrophenyl)methyl)amino)-2-pyrimidinyl)amino)phenyl)-1- pyrrolidinecarboxamide (350mg, 0.68mmol) was dissolved in methanol (5 mL) and ethyl acetate (15 mL), then tin(ll) chloride dihydrate (1.0g, 4.44 mmol) was added. The resulting mixture was heated to reflux for 2hr. The reaction mixture was diluted with ethyl acetate (100 mL), then washed with 4N NaOH (60 mL) and brine (80 mL). The organic phase was dried over Na2SO , then concentrated in vacuo to afford the titled compound (288 mg).
1H NMR (400 MHz, DMSO-d6): δ/ppm = 1.76 (m, 4H), 3.28 (m, 4H), 4.47 (d, 2H), 4.93 (s, 2H), 6.35 (d, 1 H), 6.44 (m, 2H), 6.88-7.00 (m, 3H), 7.19 (d, 1H), 7.34 (t, 1 H), 7.72 (s, 1H), 7.92 (s, 1 H), 7.97 (s, 1 H), 9.05 (s, 1 H) A13
N-[3-[[5-bromo-4-[[3-[(3-thienylmethyl)amino]propyl]amino]-2- pyrimidinyl]amino]phenyl]-1 -pyrrolidinecarboxamide
To a solution of Λ/-(3-((4-((3-aminopropyl)amino)-5-bromo-2-pyrimidinyl) amino)phenyl)-1-pyrrolidinecarboxamide (1.0 g, 1.97 mmol) in THF (30 mL) was added 2-thiophenecarboxaldehyde (184 mg, 1.64 mmol, 0.8 equiv.), triethylamine (362 mg, 3.6 mmol, 1.8 equiv) and sodium triacetoxyborohydride (688 mg, 3.25 mmol, 1.6 equiv.). The resulting mixture was stirred overnight at room temperature under N2. The reaction was quenched by satuarated sodium bicarbonate (30 mL) and was extracted with ethyl acetate (3 x 30 mL). The reaction mixture was concentrated. Purification by HPLC chromatography using acetonitrile/water gave the title compound (310 mg).
1H NMR (400 MHz, DMSO): δ/ppm = 1.81 (t, 2H), 1.87 (t, 4H), 2.88 (m, 2H), 3.32 (t, 4H), 3.54 (m, 2H), 4.30 (t, 2H), 7.04 (m, 2H), 7.17 (m, 3H), 7.59 (d, 1 H), 7.92 (s, 1 H), 8.20 (s, 1 H), 8.26 (s, 1 H), 8.62 (t, 1H), 8.82 (s, 2H), 10.48 (s, 1 H)
A14 /V2-(3-amino-5-(trifluoromethyl)phenyl)-5-bromo-/V4-(2-(7H-imidazol-4- yl)ethyl)-2,4-pyrimidinediamine and /V-(3-((5-bromo-4-((2-(τΗ-imidazol-4- yl)ethyl)amino)-2-pyrimidinyl)amino)-5-(trifluoromethyl)phenyl)- ethanimidamide
To a suspension of 5-(trifluoromethyl)-1 ,3-diaminobenzene (105 mg, 0.6 mmol, 1.2 equiv.) in acetonitrile (10 mL), hydrogen chloride (4.0/W in dioxane, 0.15 mL. 0.6 mmol) and water (0.15 mL) was added 5-bromo-2-chloro-Λ/-[2-(1/-/-imidazol-4- yl)ethyl]-4-pyrimidine (150 mg, 0.5 mmol, 1 equiv.). The resulting mixture was refluxed overnight. The resulting white suspension was cooled to room temperature and concentrated. The crude residue was purified by HPLC chromatography using acetonitrile/water to afford the title compounds, /^-(S- amino-5-(trifluoromethyl)phenyl)-5-bromo-Λ/4-(2-(' /-/-imidazol-4-yl)ethyl)-2,4- pyrimidinediamine (50 mg) and /V-(3-((5-bromo-4-((2-(7f/-imidazol-4- yl)ethyl)amino)-2-pyrimidinyl)amino)-5-(trifluoromethyl)phenyl)-ethanimidamide (22 mg).
/V2-(3-amino-5-(trifluoromethyl)phenyl)-5-bromo-/V4-(2-(7H-imidazol-4- yl)ethyl)-2,4-pyrimidinediamine: H NMR (400 MHz, DMSO-d6): δ/ppm = 2.96 (t, 2H), 3.64 (t, 2H), 6.42 (s, 1 H), 7.01 (s, 1 H), 7.24 (br t, 1 H), 7.44 (d, 2H), 8.06 (s, 1H), 8.97 (s, 1H), 9.39 (s, 1 H).
W-(3-((5-bromo-4-((2-(7H-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)-5- (trifluoromethyl)phenyl)-ethanimidamide: 1H NMR (400 MHz, DMSO-d6): δ/ppm = 2.32 (s, 3H), 2.97 (m, 2H), 3.68 (m, 2H), 7.18 (s, 1H), 7.32 (m, 1 H), 7.43 (s, 1 H), 7.79 (s, 1H), 8.13 (s, 1H), 8.36 (s, 1 H), 8.71 (s, 1 H), 8.99 (s, 1 H), 9.56 (s, 1 H), 9.92 (s, 1 H), 11.34 (s, 1 H).
A15
(4R)-Λ -[3-[[5-bromo-2-[[3-(2,5-dioxo-1-imidazolidinyl)phenyl]amino]-4- pyrimidinyl]amino]propyl]-2-oxo-4-thiazolidinecarboxamide and (4R)-N-[3- [[5-bromo-2-[[3-[2,5-dioxo-3-[[(4R)-2-oxo-4-thiazolidinyl]carbonyl]-1- imidazolidinyl]phenyl]amino]-4-pyrimidinyl]amino]propyl]-2-oxo-4- thiazolidinecarboxamide
To a solution of 3-[3-[[4-[(3-aminopropyl)amino]-5-bromo-2- pyrimidinyl]amino]phenyl]-2,4-imidazolidinedione hydrogen chloride salt (6.9 g, 13.9 mmol), (-)-2-oxo-4-thiazolidinecarboxylic acid (2.5 g, 17 mmol, 1.2 equiv.) and Λ/,Λ/-diisopropylethylamine (10 mL, 57.4 mmol, 4.1 equiv.) in dimethylformamide (150 mL) was added O-(7-azabenzotriazol-1-yl)-Λ/,Λ/,Λ/',Λ/'- tetramethyluronium hexafluorophosphate (6.5 g, 17.1 mmol, 1.2 equiv.) at 0 °C. The resulting solution was warmed to room temperature and stirred overnight. The reaction mixture was concentrated under reduced pressure to remove dimethylformamide. The crude residue was triturated in water to give a suspension. The suspension was filtered and the filter cake was washed with water and air-dried (ca. 8 g). The solid was purified by HPLC chromatography using acetonitrile/water to afford the title compounds, (4R)-Λ/-[3-[[5-bromo-2-[[3- (2,5-dioxo-1-imidazolidinyl)phenyl]amino]-4-pyrimidinyl]amino]propyl]-2-oxo-4- thiazolidinecarboxamide (2.8 g) and (4R)-N-[3-[[5-bromo-2-[[3-[2,5-dioxo-3-[[(4R)- 2-oxo-4-thiazolidinyl]carbonyl]-1-imidazolidinyl]phenyl]amino]-4- pyrimidinyl]amino]propyl]-2-oxo-4-thiazolidinecarboxamide (72 mg).
Λ/-[3-[[5-bromo-2-[[3-(2,5-dioxo-1-imidazolidinyl)phenyl]amino]-4- pyrimidinyl]amino]propyl]-2-oxo-4-thiazolidinecarboxamide: 1H NMR (400 MHz, DMSO-de): δ/ppm = 1.71 (m, 2H), 3.14 (m, 2H), 3.36 (m, 1H), 3.42 (m, 2H), 3.64 (t, 1 H), 4.04 (s, 2H), 4.23 (m, 1 H), 6.99 (d, 1H), 7.01 (t, 1 H), 7.59 (d, 1H), 7.72 (s, 1 H), 7.81 (br s, 1H), 8.16 (m, 2H), 8.29 (s, 1H), 8.34 (s, 1 H), 9.99 (br s, 1 H).
(4R)-N-[3-[[5-bromo-2-[[3-[2,5-dioxo-3-[[(4R)-2-oxo-4-thiazolidinyl]carbonyl]- 1-imidazolidinyl]phenyl]amino]-4-pyrimidinyl]amino]propyl]-2-oxo-4- thiazolidmecarboxamide: 1H NMR (400 MHz, DMSO-d6): δ/ppm = 1.64 (m, 2H), 3.12 (m, 2H), 3,38 (m, 4H), 3.79 (m, 2H), 4.02 (s, 2H), 5.04 (d, 2H), 5.12 (d, 2H), 6.94 (d, 1 H), 7.34 (t, 1H), 7.56 (d, 1H), 7.69 (s, 1H), 8.08 (s, 1 H), 8.18 (s, 1 H), 8.26 (s, 1 H), 8.37 (s, 1 H), 9.79 (br s, 1 H).
Scheme 16
Figure imgf000093_0001
Where R1, R2 and R5 are as described in the claims.
Scheme 17
Figure imgf000093_0002
Where R1, R2 and R5 are as described in the claims.
Scheme 18
Figure imgf000093_0003
Where R is C1-C4 Alkyl and R1, R2 and R5 are as described in the claims. Scheme 19
Figure imgf000094_0001
,1 Where R , R and R are as described in the claims. R -.8 and R are as described in the claims but not representing -R10.
Schema 19a
Figure imgf000095_0001
30
35 Scheme 20
Figure imgf000096_0001
SCI / Imidazole
Figure imgf000096_0002
The following Examples have been synthesized according to the above mentioned schemes. A16 V-[3-[[(2R)-2-Amino-1-oxo-3-phenylpropyl]amino]-5-[[5-bromo-4-(prop-2- ynyloxy)pyrimidin-2-yl]amino]phenyl]pyrrolidine-1-carboxamide
16a) Methyl 3-amino-5-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino] benzoate
A mixture of 5-bromo-2-chloro-4-(prop-2-ynyloxy)pyrimidine (15 g), methyl 3,5- diaminobenzoate (45 g) and concentrated hydrochloric acid (15 ml) in methanol (600 ml) was stirred at 65°C for 8 h. After concentration to half the volume water was added and the precipitate collected by filtration. The precipitate then was treated with sodium hydroxide solution (1 n) and dichloromethane. The organic phase then was washed with water and brine, dried (Na2SO ) and evaporated to dryness to give the title compound (13.8 g). Mp.: 207.5-209 °C
16b) Methyl 5-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-3-[[(2 ?)-2-
[[(1,1-dimethylethoxy)carbonyl]amino]-1-oxo-3-phenylpropyl] amino]benzoate
Λ/-BOC-D-phenylalanine (3.3 g), 1-hydroxy-1/-/-benzotriazole hydrate(1.9 g) and Λ/-[3-(dimethylamino)propyl]-/V-ethylcarbodiimid hydrochloride (2.37 g) were stirred in DMF (30 ml) for 30 minutes. Then methyl 3-amino-5-[[5-bromo-4- (prop-2-ynyloxy)pyrimidin-2-yl]amino]benzoate (3.88 g) were added and the mixture stirred over night. Then ethyl acetate (500 ml) was added and the reaction mixture washed subsequently with hydrochloric acid (0.1 n), saturated NaHCO3-solution, water and brine. After drying (Na2SO4) the organic phase was evaporated and the residue subjected to column chromatography (ethyl acetate/dichloromethane) to yield 5.36 g of the title compound. ESI-MS: 624 and 626 (M+) 16c) 5-[[5-Bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-3-[[(2/?)-2-[[(1,1- dimethylethoxy)carbonyl]amino]-1-oxo-3-phenylpropyl]amino]benzoic acid
Methyl5-[[5-bromo-4-(ρrop-2-ynyloxy)pyrimidin-2-yl]amino]-3-[[(2f?)-2-[[(1 , 1 - dimethylethoxy)carbonyl]amino]-1-oxo-3-phenylpropyl]amino]benzoate (1.0 g) was stirred in a mixture of tetrahydrofuran (20 ml), methanol (20 ml)and sodium hydroxide solution (2 n; 20 ml) for 48 h. After evaporation water (50 ml) was added to the residue. On neutralisation with hydrochloric acid (1 n) a precipitate formed. The precipitate was subjected to chromatography on silica gel (hexanes/ethyl acetate/methanol) to yield the title compound (450 mg). ESI-MS: 610 and 612 (M+)
16d) 1,1 -Dimethylethoxy [(1 ?)-2-[[3-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-
2-yl]amino]-5-[[(pyrrolidin-1-yl)carbonyl]amino]phenyl]amino]-2-oxo-1-
(phenylmethyl)ethyl]carbamate 5-[[5-Bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-3-[[(2R)-2-[[(1 ,1- dimethylethoxy)carbonyl]amino]-1 -oxo-3-phenylpropyl]amino]benzoic acid (200 mg), diphenylphosphorylazide (0.75 ml) and triethylamine (0.67 ml) were refluxed in toluene (40 ml) for 1.5 h. Then pyrrolidine (0.26 ml) was added and the mixture refluxed for additional 2 h. After cooling the reaction mixture was diluted with ethyl acetate (50 ml) and subsequently washed with saturated NaHCO3-solution, water and brine. After drying (Na2SO4) and evaporation the residue was subjected to chromatography on silica gel (hexanes/ethyl acetate) to yield the title compound (126 mg). ESI-MS: 678 and 680 (M+)
16e) Λ/-[3-[[(2 ?)-2-Amino-1 -oxo-3-phenylpropyl]amino]-5-[[5-bromo-4-(prop-2- ynyloxy)pyrimidin-2-yl]amino]phenyl]pyrrolidine-1-carboxamide
1 , 1 -Dimethylethoxy [(1 R)-2-[[3-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2- yl]amino]-5-[[(pyrrolidin-1-yl)carbonyl]amino]phenyl]amino]-2-oxo-1- (phenylmethyl)ethyl]carbamate (105 mg) and sulfuric acid (0.5 ml; 2 n) were stirred in dioxane (5 ml) at 85°C for 3.5 h. After cooling and dilution with water saturated NaHCO3-solution was added and the resulting precipitate collected by filtration yielding the title compound (76 mg). ESI-MS: 578 and 580 (M+)
A17
(αR)-α-Amino-N-[3-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-5- (hydroxymethyl)phenyl]benzenepropanamide
17a) 1 ,1 -Dimethylethoxy [(1 A?)-2-[[3-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-
2-yl]amino]-5-(hydroxymethyl)phenyl]amino]-2-oxo-1-
(phenylmethyl)ethyl]carbamate To a mixture of 5-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-3-[[(2 -?)-2- [[(1 ,1-dimethylethoxy)carbonyl]amino]-1-oxo-3-phenylpropyl]amino]benzoic acid (100 mg) and triethylamine (25 μl) in tetrahydrofuran (2 ml) was added ethyl chloroformiate (16 μl) at -10°C. After stirring for 15 minutes at 0°C sodium borohydride (19 mg) and methanol (1.6 ml) were added and stirring continued over night at room temperature. After dilution with water the reaction mixture was extracted with ethyl acetate and the organic layer subsequently washed with saturated NaHCO3-solution and brine. After drying (Na2SO4) and evaporation the residue was subjected to chromatography on silica gel (hexanes/ethyl acetate) to yield the title compound (40 mg). ESI-MS: 596 and 598 (M+)
17b) (αf?)-α-Amino-Λ/-[3-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]- 5-(hydroxymethyl)phenyl]benzenepropanamide
1 ,1-Dimethylethoxy[(1f?)-2-[[3-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino] -5-(hydroxymethyl)phenyl]amino]-2-oxo-1-(phenylmethyl)ethyl]carbamate
(22mg) and sulfuric acid (0.3 ml; 2 n) were stirred in dioxane (3 ml) at 100°C for 2.5 h. After cooling and dilution with water saturated NaHCO3-solution was added and the resulting preticipate collected by filtration yielding the title compound (10 mg). A18
3-[[5-Bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-5-[(2-hydroxyethyl) amino]benzenemethanol
18a) Methyl3-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-[(2hydroxy ethyl)amino]benzoate
Methyl3-amino-5-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]benzoate (2 g), glycolaldehyde dimer (0.7 g), sodium cyanoborohydride (0.49 g) and acetic acid (0.3 ml) were stirred in methanol (100 ml) for 24 h. After evaporation halfconcentrated NaHCO3-solution and ethyl acetate were added to the residue. The organic layer then was washed with water and brine, dried (Na2SO ), filtered and evaporated. The residue was chromatographed on silica gel (dichloromethane/methanol)to yield the title compound (1.1 g). ESI-MS: 421 and 423 (M+) Mp.: 179-179.5°C
18b) 3-[[5-Bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-5-[(2- hydroxyethyl)amino]benzoic acid
Methyl3-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-5-[(2- hydroxyethyl)amino]benzoate (350 mg) in a mixture of tetrahydrofuran (6 ml) and sodium hydroxide solution (2 n; 6 ml) was stirred for 48 h at room temperature. After evaporation the residue was diluted with water and acidified until the product precipitated. Filtration and drying yielded the title compound (340 mg). MS: 406 and 408 (M+)
18c) 2-[3-(5-Bromo-4-prop-2-ynyloxy-pyrimidin-2-ylamino)-5-hydroxymethyl- phenylamino]-ethanol
To a mixture of 3-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-5-[(2 hydroxyethyl)amino]benzoic acid and triethylamine (57 μl) in tetrahydrofuran (4 ml) was added ethyl chloroformiate (37 μl) at -10°C. After stirring for 15 minutes at 0°C sodium borohydride (44 mg) and methanol (3.6 ml) were added and stirring continued over night at room temperature. After dilution with water the reaction mixture was extracted with ethyl acetate and the organic layer subsequently washed with saturated NaHCO3-solution and brine. After drying (Na2SO ) and evaporation the residue was subjected to chromatography on silica gel (hexanes/ethyl acetate) to yield the title compound (59 mg). CI-MS: 393 and 395 (M+)
A19
Phenylmethyl[3-[[2-[[3-[[(ethylamino)carbonyl]amino]phenyl]amino]-5-
(hydroxymethyl)pyrimidin-4-yl]amino]propyl]carbamate
19a) 1-Methylethyl 2,4-dichloropyrimidine-5-carboxylate
To a precooled solution (-40°C) of 2,4-dichloropyrimidine-5-carbonyl chloride (5 ml) in tetrahydrofuran (20 ml) isopropanol (2.6 ml) was added dropwise. Then the reaction mixture was allowed to come to room temperature and stirred for 2h. After evaporation the residue was chromatographed on silica gel (dichloromethane/ethyl acetate) to yield the title compound (8.2 g).
1H NMR (300 MHz, CDCI3): σ/ppm = 1.40 (d, 6H, J = 7 Hz), 5.31 (m, 1 H), 9.0 (s, 1H)
19b) 1 -Methylethyl2-chloro-4-[[3-[[(phenylmethoxy)carbonyl]amino] propyl]amino]pyrimidine-5-carboxylate
To a solution of 1-methylethyl 2,4-dichloropyrimidine-5-carboxylate (4.7 g) and ethyldiisopropylamine (3.4 ml) in acetonitrile (250 ml) phenylmethyl [3- aminopropyl]carbamate (4.2 g) was added at 0°C. Subsequently the reaction mixture was stirred over night at room temperature. After evaporation the residue was chromatographed on silica gel (dichloromethane/isopropanol) to yield the title compound (5.9 g). ESI-MS: 407 and 409 (M+)
19c) 1 -Methylethyl2-[(3-nitrophenyl)amino]-4-[[3-[[(phenylmethoxy) carbonyl]amino]propyl]amino]pyrimidine-5-carboxylate
1-Methylethyl2-chloro-4-[[3-[[(phenylmethoxy)carbonyl]amino]propyl] amino]pyrimidine-5-carboxylate (3 g) and 3-nitroaniline (1 g) were added to a mixture of dioxane (150 ml) and hydrochloric acid in dioxane (4 n; 25 ml). After stirring at 85°C for 3.5 h the reaction mixture was poured into halfconcentrated NaHCO3-solution. The title compound precipitated and was isolated by filtration (3.5 g). ESI-MS: 509 (M+)
19d) Phenylmethyl [3-[[5-(hydroxymethyl)-2-[(3-nitrophenyl)amino] pyrimidin-4-yl]amino]propyl]carbamate
To a solution of 1-Methylethyl 2-[(3-nitrophenyl)amino]-4-[[3-[[(phenylmethoxy) carbonyl]amino]propyl]amino]pyrimidine-5-carboxylate (1.7 g) in tetrahydrofuran (100 ml) LiAIH4 (410 mg) was added in portions at 0°C. After 6h at 0°C the reaction was quenched by addition of saturated ammonium chloride solution. Ethyl acetate was added and the mixture filtered. After evaporation of the filtrate the residue was partitioned between water and dichloromethane. The organic layer was washed with brine, dried (Na2SO ), filtered and evaporated. Chromatography of the residue on silica gel (dichloromethane/methanol)) yielded the title compound (650 mg). ESI-MS: 453 (M+)
19e) Phenylmethyl [3-[[5-[[[(1 ,1 -dimethylethyl)dimethylsilyl]oxy]methy l]-2- [(3-nitrophenyl)amino]pyrimidin-4-yl]amino]propyl]carbamate
A DMF solution (5 ml) of phenylmethyl [3-[[5-(hydroxymethyl)-2-[(3- nitrophenyl)amino]pyrimidin-4-yl]amino]propyl]carbamate (250 mg), chloro(1 ,1- dimethylethyl)dimethylsilane (190 mg) and 1ry-imidazole (170 mg) was stirred at room temperature (48 h). After addition of ice water the mixture was extracted with ethyl acetate. The organic layer was washed with water, brine, dried (Na2SO ), filtered and evaporated. Trituration of the residue with diethyl ether yielded the title compound (300 mg). ESI-MS: 567 (M+)
19f) Phenylmethyl[3-[[2-[(3-aminophenyl)amino]-5-[[[(1,1-dimethylethyl) dimethylsilyl]oxy]methyl]pyrimidin-4-yl]amino]propyl]carbamate
Phenylmethyl[3-[[5-[[[(1 ,1-dimethylethyl)dimethylsilyl]oxy]methyl]-2-[(3- nitrophenyl)amino]pyrimidin-4-yl]amino]propyl]carbamate (244 mg), dissolved in ethanol (30ml), was slowly added to a mixture of FeSO4 heptahydrate (1.25 g), concentrated ammonia solution (25%; 1.25 ml) and water (5 ml). After refluxing for 3 h the mixture was filtered and the filter cake washed with ethyl acetate. The filtrate was washed with water and brine, dried (Na2SO ), filtered and evaporated to yield the crude title compound (230 mg), which was used in the next step without further purification.
19g) Phenylmethyl [3-[[5-[[[(1 ,1 -dimethylethyl)dimethylsilyl]oxy]methyl]-2- [[3-[[(ethylamino)carbonyl]amino]phenyl]amino]pyrimidin-4- yl]amino]propyl]carbamate
To a solution of phenylmethyl [3-[[2-[(3-aminophenyl)amino]-5-[[[(1 ,1- dimethylethyl)dimethylsilyl]oxy]methyl]pyrimidin-4-yl]amino]propyl]carbamate (225 mg) in acetonitrile (5 ml) ethyl isocyanate (33 μl) was added and the mixture stirred for 18 h at room temperature. Then 5 drops of ammonia solution (25%) were added and the precipitated title compound isolated by filtration (158 mg). ESI-MS: 608 (M+)
19h) Phenylmethyl [3-[[2-[[3-[[(ethylamino)carbonyl]amino]phenyl]amino]-5- (hydroxymethyl)pyrimidin-4-yl]amino]propyl]carbamate
Phenylmethyl[3-[[5-[[[(1 ,1-dimethylethyl)dimethylsilyl]oxy]methyl]-2-[[3-
[[(ethylamino)carbonyl]amino]phenyl]amino]pyrimidin-4- yl]amino]propyl]carbamate (145 mg) were stirred in a mixture of ethanol (10 ml) and hydrochloric acid (4 n; 1 ml) for 3 h at room temperature. Then halfconcentrated NaHCO3-solution and ethyl acetate were added.
The organic phase was washed with water and brine, dried (Na2SO4), filtered and evaporated to yield the title compound (120 mg).
ESI-MS: 494 (M+) 20A
1-(4-{5-Bromo-4-[2-(3H-imidazol-4-yl)-ethylamino]-pyrimidin-2-ylamino}- phenyl)-3-cyclopropyl-thiourea
20a) 2,2,2-TrifluoroN-(4-nitro-phenyl)-acetamide
4-Nitroaniline (50 g) was dissolved in pyridine (500 ml) and cooled to 0°C. Trifluoroacetic acid anhydride (52.2 ml) was added slowly at 0°C and allowed to stir at room temperature overnight. The pyridine was distilled off under reduced pressure and the solid partitioned between ethyl acetate and water. The organic phase was seperated, dried over magnesium sulfate and the solvent was removed. The crude product was recrystallized from diisopropyl ether to yield 82 g (97 %) of 2,2,2-Trifluoro-N-(4-nitro-phenyl)-acetamide which was directly used without purification in the next step.
20b) 2,2,2-TrifluoroN-(4-amino-phenyl)-acetamide
2,2,2-Trifluoro-N-(4-nitro-phenyl)-acetamide (30 g) was dissolved in ethyl acetate (500 ml) and Pd/C (10%, 3 g) was added. After hydrogenation (1bar, room temperature) for 3 h the catalyst was filtered off and the solvent was removed under reduced pressure. The crude product was recrystallized from diisopropyl ether to yield 20.6 g (79%) of 2,2,2-TrifluoroN-(4-amino-phenyl)- acetamide. ESI-MS: 205.
20c) N-(4-{5-Bromo-4-[2-(3H-imidazol-4-yl)-ethylamino]-pyrimidin- 2ylamino}-phenyl)-trifluoro acetamide 5-Bromo-4-[2-(1H-imidazol-4-yl)-ethylamino-2-chloro pyrimidine (5g, prepared according to procedure 1b) was dissolved in acetonitrile (100ml), 2,2,2-TrifluoroN- (4-amino-phenyl)-acetamide (3.37 g) and a solution of HCI in dioxane (4 M, 10 ml) were added and the reaction mixture was heated under reflux overnight. The reaction was cooled to room temperature and the precipitate was filtered and washed with acetonitrile. Yield of N-(4-{5-Bromo-4-[2-(3H-imidazol-4-yl)- ethylamino]-pyrimidin-2-ylamino}-phenyl)-trifluoro acetamide: 7.6 g (90 %). ESI- MS: 471. 20d) N2-(4-Amino-phenyl)-5-bromo-N4-[2-(3H-imidazol-4-yl)-ethyl]-pyrim idine-2,4-diamine
N-(4-{5-Bromo-4-[2-(3H-imidazol-4-yl)-ethylamino]-pyrimidin-2-ylamino}-phenyl)- trifluoro acetamide (1g, 1.9 mmole) was dissolved in THF (10 ml), MeOH (10 ml) and water (5 ml) and LiOH (455 mg) was added in one portion at room temperature. The reaction mixture was stirred at room temperature for two days, the solvent removed under reduced pressure. The residue was dissolved in ethyl acetate and water and extracted with ethyl acetate (3x). The combined organic layers were combined and dried over magnesium sulfate. After evaporation of the solvent one obtains 350 mg of N2-(4-Amino-phenyl)-5- bromo-N4-[2-(3H-imidazol-4-yl)-ethyl]-pyrimidine-2,4-diamine. ESI-MS: 375.
20e) 1-(4- 5-Bromo-4-[2-(3H-imidazol-4-yl)-ethylamino]-pyrimidin-2- ylamino}-phenyl)-3-cyclopropyl-thiourea Cyclopropyl amine (0.275 mmole) was dissolved in THF (2 ml) and thiocarbonyl diimidazole (0.28 mmole) was added. The reaction was stirred at room temperature overnight and N2-(4-Amino-phenyl)-5-bromo-N4-[2-(3H-imidazol-4- yl)-ethyl]-pyrimidine-2,4-diamine (0.26 mmole) was added as a solution in THF (3 ml) and DMF (1ml) and the reaction was stirred overnight. After removal of the solvents under reduced pressure the crude product was purified by flashmaster chromatography (dichloromethane : MeOH 9 :1) to yield 12.5 mg of 1-(4-{5-Bromo-4-[2-(3H-imidazol-4-yl)-ethylamino]-pyrimidin-2-ylamino}-phenyl)- 3-cyclopropyl-thiourea. ESI-MS: 474.
Scheme 21
Figure imgf000106_0001
Scheme 22
Figure imgf000107_0001
The following Examples have been synthesized according to the above mentioned schemes.
A21
1-(4-{5-Bromo-4-[2-(3H-imidazol-4-yl)-ethylamino]-pyrimidin-2-ylamino}- phenyl)-3-cyclopropyl-thiourea
21a) 2,2,2-TrifluoroN-(4-nitro-phenyl)-acetamide
4-Nitroaniline (50 g) was dissolved in pyridine (500 ml) and cooled to 0°C. Trifluoroacetic acid anhydride (52.2 ml) was added slowly at 0°C and allowed to stir at room temperature overnight. The pyridine was distilled off under reduced pressure and the solid partitioned between ethyl acetate and water. The organic phase was seperated, dried over magnesium sulfate and the solvent was removed. The crude product was recrystallized from diisopropyl ether to yield 82 g (97 %) of 2,2,2-Trifluoro-N-(4-nitro-phenyl)-acetamide which was directly used without purification in the next step.
21b) 2,2,2-TrifluoroN-(4-amino-phenyl)-acetamide
2,2,2-Trifluoro-N-(4-nitro-phenyl)-acetamide (30 g) was dissolved in ethyl acetate (500 ml) and Pd/C (10%, 3 g) was added. After hydrogenation (1bar, room temperature) for 3 h the catalyst was filtered off and the solvent was removed under reduced pressure. The crude product was recrystallized from diisopropyl ether to yield 20.6 g (79%) of 2,2,2-TrifluoroN-(4-amino-phenyl)- acetamide. ESI-MS: 205.
21c) N-(4-{5-Bromo-4-[2-(3H-imidazol-4-yl)-ethylamino]-pyrimidin-2-ylamino}- phenyl)-trifluoro acetamide 5-Bromo-4-[2-(1H-imidazol-4-yl)-ethylamino-2-chloro pyrimidine (5g, prepared according to procedure 1b) was dissolved in acetonitrile (100ml), 2,2,2-TrifluoroN- (4-amino-phenyl)-acetamide (3.37 g) and a solution of HCI in dioxane (4 M, 10 ml) were added and the reaction mixture was heated under reflux overnight. The reaction was cooled to room temperature and the precipitate was filtered and washed with acetonitrile. Yield of N-(4-{5-Bromo-4-[2-(3H-imidazol-4-yl)- ethylamino]-pyrimidin-2-ylamino}-phenyl)-trifluoro acetamide: 7.6 g (90 %). ESI- MS: 471. 21d) N2-(4-Amino-phenyl)-5-bromo-N4-[2-(3H-imidazol-4-yl)-ethyl]- pyrimidine-2,4-diamine
N-(4-{5-Bromo-4-[2-(3H-imidazol-4-yl)-ethylamino]-pyrimidin-2-ylamino}-phenyl)- trifluoro acetamide (1g, 1.9 mmole) was dissolved in THF (10 ml), MeOH (10 ml) and water (5 ml) and LiOH (455 mg) was added in one portion at room temperature. The reaction mixture was stirred at room temperature for two days, the solvent removed under reduced pressure. The residue was dissolved in ethyl acetate and water and extracted with ethyl acetate (3x). The combined organic layers were combined and dried over magnesium sulfate. After evaporation of the solvent one obtains 350 mg of N2-(4-Amino-phenyl)-5- bromo-N4-[2-(3H-imidazol-4-yl)-ethyl]-pyrimidine-2,4-diamine. ESI-MS: 375.
21 e) 1-(4-{5-Bromo-4-[2-(3H-imidazol-4-yl)-ethylamino]-pyrimidin-2-ylamino}- phenyl)-3-cyclopropyl-thiourea Cyclopropyl amine (0.275 mmole) was dissolved in THF (2 ml) and thiocarbonyl diimidazole (0.28 mmole) was added. The reaction was stirred at room temperature overnight and N2-(4-Amino-phenyl)-5-bromo-N4-[2-(3H-imidazol-4- yl)-ethyl]-pyrimidine-2,4-diamine (0.26 mmole) was added as a solution in THF (3 ml) and DMF (1ml) and the reaction was stirred overnight. After removal of the solvents under reduced pressure the crude product was purified by flashmaster chromatography (dichloromethane : MeOH 9 :1) to yield 12.5 mg of 1-(4-{5-Bromo-4-[2-(3H-imidazol-4-yl)-ethylamino]-pyrimidin-2-ylamino}-phenyl)- 3-cyclopropyl-thiourea. ESI-MS: 474.
A21A
1-(4-{5-Bromo-4-[2-(3-imidazol-4-yl)-ethylamino]-pyrimidin-2-ylamino}- phenyl)-3-cyclopropyl-urea
Cyclopropyl amine (0.275 mmole) was dissolved in THF (2 ml) and carbonyl diimidazole (0.28 mmole) was added. The reaction was stirred at room temperature overnight and N2-(4-Amino-phenyl)-5-bromo-N4-[2-(3H-imidazol-4- yl)-ethyl]-pyrimidine-2,4-diamine (0.26 mmole, prepared according to procedure 21) was added as a solution in THF (3 ml) and DMF (1ml) and the reaction was stirred overnight, After removal of the solvents under reduced pressure the crude product was purified by flashmaster chromatography (dichloromethane : MeOH 9 :1) to yield 23 mg (19 %) of 1-(4-{5-Bromo-4-[2-(3-imidazol-4-yl)- ethylamino]-pyrimidin-2-ylamino}-phenyl)-3-cyclopropyl-urea. ESI-MS: 458.
A22
5-Bromo-N2-(4-butylamino-phenyl)- N4-[2-(3H-imidazol-4-yl)-ethyl]- pyrimidine-2,4-diamine
N2-(4-Amino-phenyl)-5-bromo-N4-[2-(3H-imidazol-4-yl)-ethyl]-pyrimidine-2,4- diamine (1 g, 2.6 mmole, prepared according to procedure 21) was dissolved in MeOH (10 ml), butanal (0.261 ml, 2.9 mmole) was added at room temperature and the reaction mixture was stirred at room temperature for 20 minutes. Sodium cyanoborohydride (266 mg, 3.6 mmole) was added and the reaction mixture was stirred at room temperature overnight. After extraction with ethylacetate / bicarbonate solution (3x) the combined organic layers were washed with saturated NaCI-solution, dried over magnesium sulfate and evaporated. The crude product was purified by flashmaster chromatography (dichloromethane : MeOH 95:5) to provide 5-Bromo-N2-(4-butylamino-phenyl)- N4-[2-(3H-imidazol-4-yl)-ethyl]-pyrimidine-2,4-diamine (130 mg). ESI-MS: 431.
A23
N-(4-{5-Bromo-4-[2-(3H-imidazol-4-yl)-ethylamino]-pyrimidin-2-ylamino}- phenyl)-4-methanesulfonyl-3-nitro-benzamide
23a) 4-Methylsulfanyl-3-nitro-benzoic acid
4-Chloro-3-nitrobenzoic acid (10 g) were suspended in ethanol (50 ml) and water (50 ml) and sodium bicarbonate (4.16 g) was added in portions. The reaction mixture was heated at reflux for 5 minutes and NaSMe (6.95 g) was added in one portion at this temperature. The reaction was stirred under reflux for further 3 hours and then cooled to ambient temperature. The precipitate was collected by filtration to provide 4-Methylsulfanyl-3-nitro-benzoic acid (11 g, quantitative). This material was used without further purification for the following step (procedure 23b)
23b) 4-Methanesulfonyl-3-nitro-benzoic acid 4-Methylsulfanyl-3-nitro-benzoic acid (1 g, 4.69 mmole) was dissolved in methanol (25 ml) and cooled to 5°C. A solution of Oxone® (5.8 g) in water (20 ml) was added portionwise at the same temperature. The reaction mixture was allowed to stir overnight at ambient temperature, methanol was removed under reduced pressure. The suspension was diluted with water and the solid was filtered off and dried in vacuum to provide 4-Methanesulfonyl-3-nitro-benzoic acid in 89% yield (960 mg). ESI-MS: 246.
23c) N-(4-{5-Bromo-4-[2-(3H-imidazol-4-yl)-ethylamino]-pyrimidin-2- ylamino}-phenyl)-4-methanesulfonyl-3-nitro-benzamide 4-Methanesulfonyl-3-nitro-benzoic acid (72 mg, 0.29mmole) was dissolved in DMA (3 ml) and thionyl chloride (0.29 mmole) was added at ambient temperature. After the mixture was stirred for 5 minutes N2-(4-Amino-phenyl)-5- bromo-N4-[2-(3H-imidazol-4-yl)-ethyl]-pyrimidine-2,4-diamine (100 mg, 0.26 mmole, prepared according to procedure 21) was added and the reaction was allowed to stir overnight. After extraction with bicarbonate solution and ethyl acetate (3x) the combined organic layers were dried over magnesium sulfate and the solvent was removed under reduced pressure. The crude product was purified by flashmaster chromatography on silica gel to provide 37 mg of N-(4- {5-Bromo-4-[2-(3H-imidazol-4-yl)-ethylamino]-pyrimidin-2-ylamino}-phenyl)-4- methanesulfonyl-3-nitro-benzamide (23 % yield). ESI-MS: 602.
A 24
[4-(5-Bromo-4-prop-2-ynylamino-pyrimidin-2-ylamino)-phenyl]-carbamic acid butyl ester
N2-(4-Amino-phenyl)-5-bromo-N4-prop-2-ynyl-pyrimidine-2,4-diamine (0.31 mmol, prepared in analogy to procedure 21) was dissolved in THF (20 ml), triethyl amine (0.33 mmole) and butyl chloroformate (0.33 mmole) were added at room temperature and the reaction was stirred at this temperature until the starting material disappeared (TLC, 3h). The reaction was poured into water and [4-(5-Bromo-4-prop-2-ynylamino-pyrimidin-2-ylamino)-phenyl]-carbamic acid butyl ester was isolated by filtration. Yield: 91 mg (70 %). ESI-MS: 419.
A25
1-Allyl-3-[4-(5-bromo-4-prop-2-ynylamino-pyrimidin-2-ylamino)-phenyl]- thiourea
N2-(4-Amino-phenyl)-5-bromo-N4-prop-2-ynyl-pyrimidine-2,4-diamine (100 mg, 0.3 mmole, prepared in analogy procedure 21) was dissolved in acetonitrile (10 ml) and allyl isothiocyanate (1 ml) was added at room temperature. The reaction mixture was heated under reflux for 3 hours, the solvent removed under reduced pressure and 1-Allyl-3-[4-(5-bromo-4-prop-2-ynylamino-pyrimidin-2- ylamino)-phenyl]-thiourea was crystallized from acetone / ethyl acetate / hexanes. Yield 37 mg. ESI-MS: 418.
A26
1-[4-(5-Bromo-4-prop-2-ynylamino-pyrimidin-2-ylamino)-phenyl]-3-ethyl- urea
N2-(4-Amino-phenyl)-5-bromo-N4-prop-2-ynyl-pyrimidine-2,4-diamine (100 mg, 0.3 mmole, prepared in analogy to procedure 21) was dissolved in acetonitrile (10 ml) and ethyl isocyanate (0.5 ml) was added at room temperature. The reaction mixture was heated under reflux for 5 hours and then cooled to room temperature and stirred overnight. The solid was filtered off and dried under high vaccum to provide 47 mg of 1-[4-(5-Bromo-4-prop-2-ynylamino-pyrimidin-2- ylamino)-phenyl]-3-ethyl-urea. ESI-MS: 390. A27
1 -Methyl-1 H-imidazole-4-sulfonic acid [4-(5-bromo-4-prop-2-ynylamino- pyrimidin-2-ylamino)-phenyl]-amide
N2-(4-Amino-phenyl)-5-bromo-N4-prop-2-ynyl-pyrimidine-2,4-diamine (100 mg, 0.3 mmole, prepared in analogy to procedure 21) was dissolved in acetonitrile (10 ml) and triethylamine (1 ml) and 1 -Methyl-1 H-imidazole-4-sulfonyl chloride (120 mg, 0.66 mmole) was added at room temperature. The reaction mixture was stirred under reflux for 5 hours, the solvent was removed under reduced pressure and the crude product was purified by column chromatography on silica gel (ethyl acetate : hexanes 1 : 1). Yield 41 mg of 1 -Methyl-1 H-imidazole- 4-sulfonic acid [4-(5-bromo-4-prop-2-ynylamino-pyrimidin-2-ylamino)-phenyl]- amide. ESI-MS: 463.
The following examples were prepared in analogy to the compounds described above.
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
Figure imgf000166_0001
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
Figure imgf000207_0001
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
Figure imgf000211_0001
Figure imgf000212_0001
Figure imgf000213_0001
Figure imgf000214_0001
Figure imgf000215_0001
Figure imgf000216_0001
Figure imgf000217_0001
Figure imgf000218_0001
Figure imgf000219_0001
Figure imgf000220_0001
Figure imgf000221_0001
Figure imgf000222_0001
Figure imgf000223_0001
Figure imgf000224_0001
Figure imgf000225_0001
Figure imgf000226_0001
Figure imgf000227_0001
Figure imgf000228_0001
Figure imgf000229_0001
Figure imgf000230_0001
Figure imgf000231_0001
Figure imgf000232_0001
Figure imgf000233_0001
Figure imgf000234_0001
Figure imgf000235_0001
Figure imgf000236_0001
Figure imgf000237_0001
Figure imgf000238_0001
Figure imgf000239_0001
Figure imgf000240_0001
Figure imgf000241_0001
Figure imgf000242_0001

Claims

Claims:
1. Compounds of general formula (I)
Figure imgf000243_0001
(I) in which
A or B in each case independently of one another represent cyano, halogen, hydrogen, hydroxy, aryl or the group -NO2, -NH2l - NR3R4, -C1-6-alkyl-NR3R4, -N(Cι-6-hydroxyalkyl)2, -NH-C(NH)-CH3, - NH(CO)-R5, -NHCOOR6, -NR7-(CO)-NR8R9, -NR7-(CS)-NR8R9, - COOR5 , -CO-NR8R9, -CONH-Cι-6-alkyl-COOH, -SO2-CH3, 4- bromo-1 -methyl-1 H-pyrazolo-3yl or represent Cι-6-alkyl optionally substituted in one or more places, the same way or differently with halogen, hydroxy, cyano or with the group -COOR5 , -CONR8R9, -NH2, -NH-SO2-CH3, - NR8R9, -NH-(CO)-R5, -NR7-(CO)-NR8R9, -SO2-NHR3, -O-(CO)-R5 or -O-(CO)-C1-6-alkyl-R5, X represents an oxygen atom or the group -NH- or -NR3R4,
R1 represents hydrogen, halogen, hydroxymethyl, d-e-alkyl, cyano or the group -COOH, -COO-iso-propyl, -NO2, -NH-(CO)-(CH2)2- COOH or -NH-(CO)-(CH2)2-COO-C1.6-alkyl, whereby the Cι-6-alkyl can optionally be substituted in one or more places, in the same way or differently with halogen, R2 represents hydrogen or the group -NH-(CO)-aryl or d-β-alkyl optionally substituted in one or more places, the same way or differently with cyano, hydroxy, aryl, heteroaryl, C3-6- heterocycloalkylring, which can optionally be interrupted with one or more nitrogen atoms, or substituted with the group -NR8R9, - NH-(CO)-NR8R9, -NH-(CO)-S-Cι-6-alkyl, -NH-(CS)-NR8R9, -NH- (CO)O-CH2-phenyl, -NH-(CO)H, -NH(CO)-R5, -NH(CO)-OR5, - (CO)-NH-NH2, -(CO)-NH-CH2-(CO)-NH2, -(CO)-NH-Cι-6-alkyl, - COOH,
Figure imgf000244_0001
whereby the aryl or the heteroaryl can optionally be substituted in one or more places, the same or differently with halogen, hydroxy, Cι-6-alkyl, -NH2, -NH-(CO)-CH2-NH2, -NO2l -(CO)-C(CH2)-C2H5, - COOR6, -COOC(CH3)3, or represents C3-alkinyl, R3 or R4 in each case independently of one another represent hydrogen or
-6-alkyl optionally substituted in one or more places, the same way or differently with hydroxy, phenyl or hydroxyphenyl, or R3 and R4 together form a C3-6-heterocycloalkylring containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C3-6- heterocycloalkylring can optionally be substituted with C1-6-alkyl, Cι-6-alkyl-COOH or C1-6-alkyl-NH2,
R5 represents hydrogen, d-β-alkyl, Cι-6-alkoxy, C2-6-alkenyl, C3-e- cycloalkylring, aryl, heteroaryl, the group -(CO)-NH2 or C3.6- heterocycloalkylring that can optionally be interrupted with one or more nitrogen and/or oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring and d-6-alkyl, C2-6-alkenyl, C3-6-cycloalkylring, C3-6- heterocycloalkylring defined above, aryl or heteroaryl can optionally be substituted in one or more places, the same way or differently with halogen, hydroxy, d-β-alkyl, Cι-6-alkoxy, C3.6- cycloalkyl, C3-6-heterocycloalkylhng defined above, aryl, heteroaryl or with the group -NR8R9, -NO2l -NR7-(CO)-R5, -NH(CO)-C1-6- alkyl-NH-(CO)-Cι-6-alkyl, -NR7-(CO)-NR8R9, -CO-CH3) -COOH, - CO-NR8R9, -SO2-aryl, -SH, -S-Cι-6-alkyl, -SO2-NR8R9, whereby aryl itself can optionally be substituted in one or more places, the same way or differently with halogen, hydroxy, Chalky! or d-β-alkoxy, R6 represents d-6-alkyl, C2-6-alkenyl or phenyl, whereby d-6-alkyl may optionally be substituted with C3-6- heterocycloalkylhng that can optionally be interrupted with one or more nitrogen and/or oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, R7 represents hydrogen or Cι-6-alkyl,
R8or R9 in each case independently of one another represent hydrogen, d-6-alkyl, C2-6-alkenyl, C3-6-cycloalkyl, aryl or heteroaryl or the group R10, whereby d-6-alkyl, C2-6-alkenyl, C3-6-cycloalkyl, aryl or heteroaryl can optionally be substituted in one or more places, the same way or differently with halogen, heteroaryl, hydroxy, d-β-alkoxy, hydroxy-d-e-alkoxy or the group -COOH, -NO2, -NR8R9, -N(d-6- alkyl)2 or with a C3-6-heterocycloalkylring can optionally be interrupted with one or more nitrogen and/or oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, or R8 and R9 together form a C3-6-heterocycloalkylring containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C3-6- heterocycloalkylring can optionally be substituted in one or more places, the same way or differently with hydroxy or the group - NR8R9, -NH(CO)-R5, hydroxy-C1-6-alkyl or -COOH and R10 represents -SO2-aryl, -SO2-heteroaryl or -SO2-NH2 or -SO2-C1-6- alkyl, whereby the aryl can be substituted with -Cι-6-alkyl, with the following provisos: whereby when X represents -NR3R4 then R2 does not represent a substituent, whereby when A and B represent hydrogen, X represents -NH- and R2 represents Cι-6-alkyl, then R1 represents -NH-(CO)-CH(NH2)-(CH2)2-COOH or -NH-
(CO)-CH(NH2)-(CH2)2-COOC2H5, whereby when A represents-(CO)-OC2H5 or hydroxy, B represents hydrogen, X represents oxygen, R1 represents halogen, then R2 represents C3-alkinyl, whereby when A represents -(CO)-OC2H5 or hydroxy, B represents hydrogen, X represents -NH-, R represents -NO2, then R2 represents C3-alkinyl, whereby when A represents -(CO)-OCH3, then X represents oxygen, R1 represents halogen, R2 represents C3-alkinyl and B represenst -NH2, -NHC2H4OH, -N(C2H4OH)2, -
NH-(CO)-CH2-O(CO)CH3, whereby when A represents -(CO)-OCH3, then X represents -NH-, R1 represents halogen, R2 represents - C2H4-imidazolyl and B represenst hydrogen -NH2> whereby when A represents -NHS02-CH3, then B represents hydrogen, X represents -NH-, R1 represents halogen and R2 represents -C2H -imidazolyl, whereby when R1 represents -COO-iso-propyl, then X represents -NH- and R2 represents C3-alkinyl and A or B independently of one another represent the group -NO2 or -NH-
(CO)-CF3, whereby when R1 represents halogen, X represents -NH-, B represents hydrogen and R2 represents d^-alky! substituted with -NH2, then A represents -NH-(CO)-C6-cycloalkyl-NH2, whereby when R1 represents halogen, X represents -NH-, B represents -S-
CH3 and R2 represents imidazolyl, then A represents the group
Figure imgf000248_0001
as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
2. Compounds of general formula (I), according to claim 1 in which A or B in each case independently of one another represent cyano, halogen, hydrogen, hydroxy, tetrazolyl or the group -NH2) -NR3R4, -Cι-6-alkyl-NR3R4, -NH-C(NH)-CH3, -NH(CO)-R5, -NHCOOR6, -
NR7-(CO)-NR8R9, - Cι-6-alkyl-COOH, -COOH , -CONH2, -CONH- C1-6-alkyl-COOH, or represent Cι-6-alkyl optionally substituted in one or more places, the same way or differently with halogen, hydroxy or with the group -COOH , -CONR8R9, -NH-SO2-CH3 or -NR8R9,
X represents the group -NH- or -NR3R4 ,
R1 represents cyano, hydrogen, halogen or C1-6-alkyl, whereby the C*.
6-alkyl can optionally be substituted in one or more places, in the same way or differently with halogen, R2 represents hydrogen or the group -NH-(CO)-aryl or -d-e-alkyl optionally substituted in one or more places, the same way or differently with cyano, hydroxy, aryl , heteroaryl, C3-6- heterocycloalkylring which can be optionally be interrupted in one or more places with one or more nitrogen atoms, or substituted with the group -NR8R9, -NH-(CO)-NR8R9, -NH-(CO)-S-C1-6-alkyl, -
NH-(CS)-NR8R9, -NH(CO)-R5, -NH(CO)-OR5, -(CO)-NH-NH2, - (CO)-NH-CH2-(CO)-NH2> -(CO)-NH-Cι-6-alkyl, -COOH whereby the aryl or the heteroaryl can optionally be substituted in one or more places, the same way or differently with hydroxy, Cι-6-alkyl, -NH2, - NH-(CO)-CH2-NH2, -NO2, -COOR6,
Figure imgf000249_0001
R3 or R4 in each case independently of one another represent hydrogen, Cι-6-alkyl optionally substituted in one or more places, the same way or differently with hydroxy, phenyl or hydroxyphenyl, or
R3 and R4 together form a C3-6-heterocycloalkylring containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C3-6- heterocycloalkylnng can optionally be substituted with d-6-alkyl, d-e-alkyl-COOH or C1-6-alkyl-NH2, R5 represents hydrogen, C1-6-alkyl, d-β-alkoxy, C2-6-alkenyl, C3-6- cycloalkylring, heteroaryl , the group -(CO)-NH2 or C3-6- heterocycloalkylnng that can optionally be interrupted with one or more nitrogen and/or oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring and d-6-alkyl, C2-6-alkenyl, C3.6-heterocycloalkylhng define above, aryl or heteroaryl can optionally be substituted in one or more places, the same way or differently with halogen, hydroxy, d-6- alkyl, Cι_6-alkoxy, C3-6-cycloalkyl, C3.6-heterocycloalkylring define above, aryl, heteroaryl or with the -NR8R9, -NO2, -NR7-(CO)-R5, - NH(CO)-C1-6-alkyl-NH-(CO)-C1-6-alkyl, -NR7-(CO)-NR8R9, -CO-CH3,
-COOH, -CO-NR8R9, -SO2-aryl, -SH, -S-C1-6-alkyl, -SO2-NR8R9, whereby aryl itself can optionally be substituted in one or more places, the same way or differently with halogen or hydroxy, C1-6- alkyl or Cι-6-alkoxy, R7 represents hydrogen or Cι-6-alkyl,
R8or R9 in each case independently of one another represent hydrogen, d-6-alkyl, aryl or heteroaryl or the group R10, whereby d-6-alkyl, aryl or heteroaryl can optionally be substituted in one or more places, the same way or differently with halogen, heteroaryl, hydroxy, Cι-6-alkoxy, hydroxy-Cι_6-alkoxy or with the group -
COOH, -NO2, or a C3-6-heterocycloalkylring can optionally be interrupted with one or more nitrogen and/or oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring or R8 and R9 together form a C3-6-heterocycloalkylring containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C3-6- heterocycloalkylring can optionally be substituted in one or more places, the same way or differently with hydroxy, hydroxy-C1-6-alkyl or the group -NR8R9, -NH(CO)-R5 or -COOH and
R10 represents -SO2-NH2, -SO2-Cι-6-alkyl, -SO2-aryl, or -SO2- heteroaryl, whereby the aryl can be substituted with -d-6-alkyl, as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
3. Compounds of general formula (I) according to claim 1 or 2 in which A or B in each case independently of one another represent hydrogen, tetrazolyl or the group -N(CH3)2, -NH-(CO)-pyrrolidinyl, -NH-(CO)- pentyl, -NH-(CO)-hexyl, -NH-(CO)-hexyl-NH2, -NH-(CO)-C3H7, - NH-(CO)-CH2-phenyl, -NH-(CO)-CH2-NH2, -NH-(CO)-C2H4-NH2, - NH-(CO)-CH(NH2)-CH3, -NH-(CO)-CH(NH2)-hydroxyphenyl, -NH- (CO)-CH(NH2)-CH2-phenyl, -NH-(CO)-CH(NH2)-CH2- hydroxyphenyl, -NH-(CO)-CH(NH-(CO)-CH3)-CH2-phenyl, -NH- (CO)-CH2-NH-(CO)-CH3, -NH-(CO)-N(C2H5)(C2H4-piperidinyl), -NH- (CO)-N(CH3)(C2H4-piperidinyl), -NH-(CO)-CH2-NH(CH3), -CH2- N(CH3)2, -NH-(CO)NH-CH2-COOH , hydantoinyl, -CH2-COOH whereby the pyrrolidinyl can optionally be substituted with hydroxy or the group -NH2, -N(CH3)2 or -NH-(CO)-CH3, and whereby hydantoinyl can be substituted with -CH3, -CH2- COOH, or -(CO)-thiazolidinonyl, X represents or the group -NH-, R1 represents halogen and
R2 represents hydrogen or the group -NH-(CO)-phenyl or -C2H -, -C3H6- both can optionally be substituted in one or more places, the same way or differently with cyano, hydroxy, phenyl, naphthyl, imidazolyl, thiazolyl, pyridyl, 2-oxazolinyl, piperidinyl, - NH2, -NH-CH2-thienyl, -NH-pyridinyl-NO2, -NH-thiazolyl, -SO2- thienyl, -SO2-NH2, -SO2-CH3, -SO2-C3H7, pyrrolidinonyl substituted with -COOH, -NH-(CO)-NH-thienyl, -NH-(CO)-NH-phenyl, -NH- (CO)-NH- C2H5, -NH-(CO)-C(CH3)3, -NH-(CO)-S-C2H5, -NH-(CS)- NH- C2H5, -NH-(CO)-C2H5, -NH-(CO)-thienyl, -(CO)-NH-NH2, - (CO)-NH-CH2-(CO)-NH2, -(CO)-NH-C2H5, -COOH whereby the phenyl or the imidazolyl, thiazolyl can optionally be substituted in one or more places, the same way or differently with hydroxy, - CH3, -NH-(CO)-CH2-NH2, -COOC2H5, -COOC(CH3)3,
Figure imgf000252_0001
as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
4. Compounds of general formula (I) according to any one of claims 1 to 3 in which
A or B in each case independently of one another represent hydrogen or the group -NH-(CO)-pyrrolidinyl, -NH-(CO)-piperidinyl, -NH-(CO)- morpholinyl, -NH-(CO)-hexyl-NH2, -NH-(CO)-CH(NH2)- hydroxyphenyl, -NH-(CO)-CH(NH2)-CH2-hydroxyphenyl, hydantoin optionally substituted with -CH3,
X represents or the group -NH-,
R1 represents halogen and
R2 represents hydrogen, -C2H4-imidazolyl or -C3H wich can optionally be substituted in one or more places, the same way or differently with the group -NH-CH2-thienyl, -NH-(CO)-C2H5, -NH-(CO)-
C(CH3)3,
Figure imgf000254_0001
as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
5. Compounds of general formula (I) according to claim 4, N-[3-[[5-bromo-4-[[3-[[[1-
(trifluoromethyl)cyclobutyl]carbonyl]amino]propyl]amino]-2- pyhmidinyl]amino]phenyl]-1-pyrrolidinecarboxamide, N-[3-[[5-bromo-4-[[3-[[1-oxo-3-(phenylsulfonyl)propyl]amino]propyl]amino]-2- pyhmidinyl]amino]phenyl]-1-pyrrolidinecarboxamide, N-[3-[[5-bromo-2-[[3-[(1-pyrrolidinylcarbonyl)amino]phenyl]amino]-4- pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide, N-[3-[[4-[[3-[[(1-aminocyclopentyl)carbonyl]amino]propyl]amino]-5-bromo-2- pyrimidinyl]amino]phenyl]-1-pyrrolidinecarboxamide, N-[3-[[4-[[3-[[(1-aminocyclobutyl)carbonyl]amino]propyl]amino]-5-iodo-2- pyrimidinyl]amino]phenyl]-1 -pyrrolidinecarboxamide,
N1-[3-[[5-bromo-2-[[3-[(1-pyrrolidinylcarbonyl)amino]phenyl]amino]-4- pyrimidinyl]amino]propyl]-1 ,1-cyclopentanedicarboxamide, (4R)-Λ/-[3-[[5-bromo-2-[[3-(2,5-dioxo-1-imidazolidinyl)phenyl]amino]-4- pyrimidinyl]amino]propyl]-2-oxo-4-thiazolidinecarboxamide, (4R)-N-[3-[[5-bromo-2-[[3-(3-methyl-2,5-dioxo-1- imidazolidinyl)phenyl]amino]-4-pyrimidinyl]amino]propyl]-2-oxo-4- thiazolidinecarboxamide,
3-[3-[[5-bromo-4-[[2-(1 H-imidazol-4-yl)ethyl]amino]-2- pyrimidinyl]amino]phenyl]-2,4-imidazolidinedione, 3-[3-[[5-bromo-4-[[2-(1 H-imidazol-4-yl)ethyl]amino]-2- pyrimidinyl]amino]phenyl]-1-methyl-2,4-imidazolidinedione, N'-[3-[[5-bromo-4-[[2-(1 H-imidazol-4-yl)ethyl]amino]-2- pyhmidinyI]amino]phenyl]-N-ethyl-N-[2-(1 -piperidinyl)ethyl]-urea, N-[3-[[5-bromo-4-[[3-[(2,2-dimethyl-1-oxopropyl)amino]propyl]amino]-2- pyrimidinyl]amino]phenyl]-1 -pyrrolidinecarboxamide,
N-[3-[[2-[[3-[[(2S)-2-amino-3-(4-hydroxyphenyl)-1- oxopropyl]amino]phenyl]amino]-5-bromo-4-pyrimidinyl]amino]propyl]-2,2- dimethyl-propanediamide, N-[3-[[2-[[3-[[(1 -aminocyclohexyl)carbonyl]amino]phenyl]amino]-5-bromo-4- pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide,
N-[3-[[2-[[3-[[(2S)-2-amino-2-phenylacetyl]amino]phenyl]amino]-5-bromo-4- pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide, N-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5- bromo-4-pyrimidinyl]amino]propyl]-5-oxo-2-pyrrolidinecarboxamide, N-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5- bromo-4-pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide, N1-[3-[[5-bromo-2-[[3-[[(2S)-2-pyrrolidinylcarbonyl]amino]phenyl]amino]-4- pyrimidinyl]amino]propyl]- 1 ,1-cyclopropanedicarboxamide, N-[3-[[5-bromo-2-[[3-(2,5-dioxo-1-imidazolidinyl)phenyl]amino]-4- pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide,
/V-(3-((5-bromo-4-((2-(7H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-4-morpholinecarboxamide, Λ/-(3-((5-bromo-4-((2-( 7H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-1-pyrrolidinecarboxamide,
Λ/-(3-((5-bromo-4-((3-((2-thienylcarbonyl)amino)propyl)amino)-2- pyrimidinyl)amino)phenyl)-1-pyrrolidinecarboxamide,
/V7-(3-((5-bromo-2-((3-((1-pyrrolidinylcarbonyl)amino)phenyl)amino)-4- pyrimidinyl)amino)propyl)-1 ,1-cyclopropanedicarboxamide,
Λ/-(3-((5-bromo-4-((3-((1-oxopropyl)amino)propyl)amino)-2- pyrimidinyl)amino)phenyl)-1 -pyrrolidinecarboxamide,
Λ/-(3-((5-iodo-4-((3-((2-thienylcarbonyl)amino)propyl)amino)-2-pyrimidinyl)- amino)phenyl)-1 -pyrrolidinecarboxamide, N-[3-[[5-bromo-4-[[3-[[[(2S)-5-oxo-2- pyrrolidinyl]carbonyl]amino]propyl]amino]-2-pyrimidinyl]amino]phenyl]-1- pyrrolidinecarboxamide,
N-[3-[[5-bromo-4-[[3-[[[(2S)-4-oxo-2-azetidinyl]carbonyl]amino]propyl]amino]-
2-pyrimidinyl]amino]phenyl]-1-pyrrolidinecarboxamide, (4R)-N-[3-[[5-bromo-2-[[3-[(1-pyrrolidinylcarbonyl)amino]phenyl]amino]-4- pyrimidinyl]amino]propyl]-2-oxo-4-thiazolidinecarboxamide or
N-[3-[[4-[[3-[[(1-aminocyclobutyl)carbonyl]amino]propyl]amino]-5-bromo-2- pyrimidinyl]amino]phenyl]-1 -pyrrolidinecarboxamide.
ompounds of general formula (I) according to claim 1 , in which
A or B in each case independently of one another represent hydrogen or the group -NO2, -NH2, -NR3R4, -N(C1-6-hydroxyalkyl)2, -NH(CO)- R5, -NHCOOR6, -NR7-(CO)-NR8R9, -NR7-(CS)-NR8R9, -COOR5, - CO-NR8R9, -SO2-CH3, 4-bromo-1 -methyl-1 H-pyrazolo-3yl or d-6-alkyl optionally substituted in one or more places, the same way or differently with cyano, halogen, hydroxy or the group -NH2, -NH-(CO)-R5, -SO2-NHR3, -COOR5, -CONR8R9, -O-(CO)-R5, -O-
(CO)-C1-6-alkyl-R5, X represents an oxygen atom or the group -NH-,
R1 represents hydrogen, halogen, hydroxymethyl or the group - COOH, -COO-iso-propyl, -NO2, -NH-(CO)-(CH2)2-COOH or -NH-
(CO)-(CH2)2-COO-Cι-6-alkyl, R2 represents Cι_6-alkyl optionally substituted in one or more places, the same way or differently with hydroxy, imidazolyl or the group -
NH2, -NH-(CO)O-CH2-phenyl, -NH-(CO)H, -NH-(CO)-phenyl, -NH- (CO)-CH2-O-phenyl, -NH-(CO)-CH2-phenyl, -NH-(CO)-
CH(NH2)CH2-phenyl, -NH-(CO)-CH2-CH(CH3)-phenyl, -NH-(CO)-
CH(NH2)-(CH2)2-COOH,
Figure imgf000257_0001
, whereby the phenyl can optionally be substituted in one or more places, the same or differently with halogen, d-β-alkyl or -(CO)-
C(CH2)-C2H5, or represents C3-alkinyl, R3 or R4 in each case independently of one another represent hydrogen or
Cι-6-alkyl optionally substituted in one or more places, the same way or differently with hydroxy, phenyl or hydroxyphenyl, or R3 and R4 together form a C3-6-heterocycloalkylhng containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupoted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C3-6- heterocycloalkylring can optionally be substituted with d-6-alkyl, d-e-alkyl-COOH or Cι-6-alkyl-NH2,
R5 represents d-β-alkyl, C2-6-alkenyl, C3-6-cycloalkyl or phenyl each can optionally be substituted in one or more places, the same way or differently with halogen, hydroxy, phenyl or with the group -NH2l -NH(CO)-O-Cι-6-alkyl, whereby phenyl itself can optionally be substituted in one or more places, the same way or differently with halogen, hydroxy or d-6-alkyl,
R6 represents d-β-alkyl, C2-6-alkenyl or phenyl,
R7 represents hydrogen or d-β-alkyl and
R8or R9 in each case independently of one another represent hydrogen, d-β-alkyl, C2-6-alkenyl, C3-6-cycloalkyl, aryl or phenyl, whereby aryl or phenyl can optionally be substituted in one or more places, the same way or differently with hydroxy or the group -NO2 or -N(C1-6- alkyl)2 or R8 and R9 together form a C3-6-heterocycloalkylring containing at least one nitrogen atom and optionally can be interrupted by one or more oxygen and/or sulfur atoms and/or can be interrupted by one or more -(CO)- groups in the ring and/or optionally can contain one or more possible double bonds in the ring, whereby the C3-6- heterocycloalkylnng can optionally be substituted with the group -
NH2, as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
ompounds of general formula (I) according to claim 1 or 6 in which
A or B in each case independently of one another represent hydrogen or the group -NH-C2H4-OH, -NH-CH2-hydroxyphenyl, -NH-(CO)- pyrrolidinyl, -NH-(CO)-CH(NH2)-CH2-phenyl, -NH-(CO)-pentyl-NH2, -NH-(CO)-hexyl-NH2, -NH-(CO)-CH2-NH2, -NH-(CO)-CH(NH2)- hydroxyphenyl, -NH-(CO)-CH2-hydroxyphenyl, -NH-(CO)-CH2- methylphenyl, -NH-(CO)-C2H4-dihydroxyphenyl, -NH-(CO)- CH(OH)-phenyl, -NH-(CO)-CH(NH2)-CH2(OH), -NH-(CO)-
C(CH3)2NH2, -NH-(CO)-NH(C2H5), -CH2OH, -(CO)-NH-cyclopropyl,
-(CO)-NH-CH(CH3)2, whereby the pyrrolidinyl can optionally be substituted with hydroxy or the group -NH2, X represents an oxygen atom or the group -NH-,
R1 represents halogen or hydroxymethyl and
R2 represents -C2H5 optionally substituted in one or more places, the same way or differently with hydroxy, imidazolyl or represents -C3H or -C4H8 optionally substituted in one or more places, the same way or differently with the group -NH2, -NH-
(CO)-CH(NH2)-C2H4-COOH, -NH-(CO)-phenyl, -NH-(CO)-CH2- phenyl, -NH-(CO)-CH2-CH(CH3)-phenyl, -NH-(CO)-CH2-O-phenyl, -NH-(CO)O-CH2-phenyl, -NH-(CO)-CH(NH2)CH2-phenyl,
Figure imgf000259_0001
whereby the phenyl can optionally be substituted in one or more places, the same or differently with halogen, -CH3 or -(CO)- C(CH2)(C2H5), or represents C3-alkinyl, as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
8. Compounds of general formula (I) according to claim 7,
N-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5- bromo-4-pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide,
1-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5- bromo-4-pyrimidinyl]amino]propyl]-2-oxo-3-pyrrolidinecarboxylic acid,
Λ/-[3-[[5-bromo-4-[[3-[[(5-oxo-2-pyrrolidinyl)carbonyl]amino]propyl]amino]-2- pyrimidinyl]amino]phenyl]-1 -pyrrolidinecarboxamide,
Pyrrolidine-1 -carboxylic acid [3-(5-bromo-4-{3-[2-(2,4-dichloro-phenyl)- acetylamino]-propylamino}-pyhmidin-2-ylamino)-phenyl]-amide, Pyrrolidine-1 -carboxylic acid [3-(5-bromo-4-{3-[2-(4-bromo-phenyl)- acetylamino]-propylamino}-pyhmidin-2-ylamino)-phenyl]-amide, Pyrrolidine-1 -carboxylic acid (3-{5-bromo-4-[3-(2-p-tolyl-acetylamino)- propylamino]-pyrimidin-2-ylamino}-phenyl)-amide, Pyrrolidine-1 -carboxylic acid [3-(5-bromo-4-{3-[2-(2,4-difluoro-phenyl)- acetylamino]-propylamino}-pyhmidin-2-ylamino)-phenyl]-amide,
Pyrrolidine-1 -carboxylic acid {3-[5-bromo-4-(3-{2-[2,3-dichloro-4-(2- methylene-butyryl)-phenoxy]-acetylamino}-propylamino)-pyrimidin-2- ylamino]-phenyl}-amide,
Pyrrolidine-1-carboxylic acid [3-(5-bromo-4-{3-[3-(2,3-dichloro-phenyl)- butyrylamino]-propylamino}-pyrimidin-2-ylamino)-phenyl]-amide,
Pyrrolidine-1 -carboxylic acid (3-{5-bromo-4-[3-(3-bromo-benzoylamino)- propylamino]-pyrimidin-2-ylamino}-phenyl)-amide, Λ/-(3-((4-((4-aminobutyl)amino)-5-bromo-2-pyhmidinyl)amino)phenyl)-1- pyrrolidinecarboxamide, N-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5- bromo-4-pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide, Λ/-[3-[[(2S)-2-Amino-1-oxo-3-phenylpropyl]amino]-5-[[5-bromo-4-(prop-2- ynyloxy)pyhmidin-2-yl]amino]phenyl]pyrrolidine-1-carboxamide, Λ -[3-[[(2 )-2-Amino-1-oxo-3-phenylpropyl]amino]-5-[[5-bromo-4-(prop-2- ynyloxy)pyrimidin-2-yl]amino]phenyl]pyrrolidine-1-carboxamide,
(αR)-α-Amino-Λ/-[3-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-5-
(hydroxymethyl)phenyl]benzenepropanamide, 2-[3-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-5-hydroxymethyl- phenylaminoj-ethanol,
(2R)-Amino-N-[3-hydroxymethyl-5-(4-prop-2-ynyloxy-pyrimidine-2-ylamino)- phenyl]-3-phenyl-propionamide,
3-((2R)-Amino-3-phenyl-propionylamino)-5-(5-bromo-4-prop-2-ynyloxy- pyrimidine-2-ylamino)- N-cyclopropyl-benzamide,
3-((2R)-Amino-3-phenyl-propionylamino)-5-(5-bromo-4-prop-2-ynyloxy- pyrimidin-2-ylamino)- N-isopropyl-benzamide,
Phenylmethyl [3-[[2-[[3-[[(ethylamino)carbonyl]amino]phenyl]amino]-5-
(hydroxymethyl)pyrimidine-4-yl]amino]propyl]carbamate, Pyrrolidine-1 -carboxylic acid (3-{4-[3-((2R)-amino-3-phenyl-propionylamino)- propylamino]-5-bromo-pyrimidine-2-ylamino}-phenyl)-amide,
Pyrrolidine-1 -carboxylic acid (3-{4-[3-((2S)-amino-3-phenyl-propionylamino)- propylamino]-5-bromo-pyrimidine-2-ylamino}-phenyl)-amide,
2-[3-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenylamino]-ethanol, 1 -Amino-cyclopentancarbonylic acid[3-(5-bromo-4-prop-2-ynyloxy- pyrimidine-2-ylamino)-phenyl]-amide,
1 -Amino-cyclohexancarbonylic acid-[3-(5-bromo-4-prop-2-ynyloxy- pyrimidine-2-ylamino)-phenyl]-amide,
(2S)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-3- phenyl-propionamide,
(2R)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-3- phenyl-propionamide,
2-{[3-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenylamino]- methyl}-phenol, (2R)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-3-
(4-hydroxy-phenyl)-propionamide,
N-[3-(5-Bromo-4-prop-2-ynyloxy-pyhmidine-2-ylamino)-phenyl]-3-(3,4- dihydroxy-phenyl)-propionamide, N-[3-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-2-hydroxy-
(2S)-phenyl-acetamide,
N-[3-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-2-hydroxy-
(2R)-phenyl-acetamide, (2S)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-3- hydroxy-propionamide,
(2R)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidin-2-ylamino)-phenyl]-3- hydroxy-propionamide,
2-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyhmidine-2-ylamino)-phenyl]-2- methyl-propionamide,
(2S)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-3-
(4-hydroxy-phenyl)-propionamide,
(2S)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-3- p-tolyl-propionamide or (2R)-Amino-N-[3-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenyl]-3- p-tolyl-propionamide.
9. Compounds of general formula (I) according to claim 1 in which A or B in each case independently of one another represent halogen, hydrogen or the group -SO2-CH3, -NO2, -NH2, -CF3, -CH2-NH- (CO)-NH2, -CH2-pyrrolidinyl, -NH-(CO)-CH3, -NH-(CO)-hexyl-NH2, - NH-(CO)-phenyl, -NH-(CO)-pyrrolidinyl, -NH-(CO)-CH(NH2)-CH2- phenyl, NH-(CO)-OCH3, -NH-(CO)-OCH(CH3)2, -NH-(CO)-OC2H4- morpholino, -NH-(CO)-NH-cyclopropyl, -NH-(CO)-morpholino, -NH-
(CO)-NH-C2H4-morpholino, -NH-(CO)-NH-hydroxycycloalkyl, hydantoinyl, whereby the pyrrolidinyl can optionally be substituted with hydroxy or the group -NH2 and whereby the hydantoinyl can optionally be substituted with the group -CH3 or -(CO)-thiazolidinonyl, X represents the group -NH-,
R1 represents halogen and R2 represents -CH2-dihydroxyphenyl, -C2H4-imidazolyl, or -C3H7 optionally substituted in one or more places, the same way or differently with
Figure imgf000263_0001
as well as all related isotopes, diastereomers, enantiomers, solvates, polymorphs or pharmaceutically acceptable salts thereof.
10. Compounds of general formula (I) according to claim 7,
4-((4-((2-(1 H-imidazol-4-yl)ethyl)amino)-5-iodo-2-pyrimidinyl)amino)- benzenesulfonamide,
N-((3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)methyl)-urea, 1-((3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)methyl)-3-pyrrolidinol, (3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-carbamic acid methyl ester, N2-(3-aminophenyl)-5-bromo-N4-(2-(1 H-imidazol-4-yl)ethyl)-2,4- pyrimidinediamine, N-(3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-N'-cyclopropyl-urea,
N-(3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-4-morpholinecarboxamide, (3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyhmidinyl)amino)phenyl)-carbamic acid 1-methylethyl ester, N-(3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-methanesulfonamide, N2-(3-amino-5-(trifluoromethyl)phenyl)-5-bromo-N4-(2-(1 H-imidazol-4- yl)ethyl)-2,4-pyrimidinediamine, N-(3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-N'-(2-(4-morpholinyl)ethyl)-urea, N2-(3-amino-5-chlorophenyl)-5-bromo-N4-(2-(1 H-imidazol-4-yl)ethyl)-2,4- pyrimidinediamine,
(3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyhmidinyl)amino)phenyl)-carbamic acid 2-(4-morpholinyl)ethyl ester, N-(3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-N'-(4-hydroxycyclohexyl)-urea, N-(3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyhmidinyl)amino)phenyl)-acetamide, N-(3-((5-bromo-4-((2-(1 H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)-benzamide, (4R)-N-[3-[[5-bromo-2-[[3-[(1-pyrrolidinylcarbonyl)amino]phenyl]amino]-4- pyrimidinyl]amino]propyl]-2-oxo-4-thiazolidinecarboxamide,
3-[3-[[5-bromo-4-[[2-(1 H-imidazol-4-yl)ethyl]amino]-2- pyrimidinyl]amino]phenyl]-2,4-imidazolidinedione, 3-[3-[[5-bromo-4-[[2-(1 H-imidazol-4-yl)ethyl]amino]-2- pyhmidinyl]amino]phenyl]-1-methyl-2,4-imidazolidinedione, 1-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5- bromo-4-pyrimidinyl]amino]propyl]-2-oxo-3-pyrrolidinecarboxylic acid, 1-[3-[[2-[[3-[[(1-aminocyclohexyl)carbonyl]amino]phenyl]amino]-5-bromo-4- pyhmidinyl]amino]propyl]-2-oxo-3-pyrrolidinecarboxylic acid, N-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5- bromo-4-pyrimidinyl]amino]propyl]-5-oxo-2-pyrrolidinecarboxamide,
N-[3-[[2-[[3-[[(2R)-2-amino-1-oxo-3-phenylpropyl]amino]phenyl]amino]-5- chloro-4-pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide, 3-[3-[[5-bromo-4-[[(3,4-dihydroxyphenyl)methyl]amino]-2- pyrimidinyl]amino]phenyl]-2,4-imidazolidinedione, 3-[3-[[5-bromo-4-[[(3,4-dihydroxyphenyl)methyl]amino]-2- pyrimidinyl]amino]phenyl]-1-methyl-2,4-imidazolidinedione, (4R)-N-[3-[[5-bromo-2-[[3-(2,5-dioxo-1-imidazolidinyl)phenyl]amino]-4- pyrimidinyl]amino]propyl]-2-oxo-4-thiazolidinecarboxamide, N-[3-[[5-bromo-2-[[3-(2,5-dioxo-1-imidazolidinyl)phenyl]amino]-4- pyrimidinyl]amino]propyl]-5-oxo-2-pyrrolidinecarboxamide, N-[3-[[5-bromo-2-[[3-(2,5-dioxo-1-imidazolidinyl)phenyl]amino]-4- pyrimidinyl]amino]propyl]-2,2-dimethyl-propanediamide, 3-[3-[[5-bromo-4-[[3-(2-oxo-1-pyrrolidinyl)propyl]amino]-2- pyrimidinyl]amino]phenyl]-2,4-imidazolidinedione,
(4R)-N-[3-[[5-bromo-2-[[3-(3-methyl-2,5-dioxo-1-imidazolidinyl)phenyl]amino]- 4-pyrimidinyl]amino]propyl]-2-oxo-4-thiazolidinecarboxamide or (4R)-N-[3-[[5-bromo-2-[[3-[2,5-dioxo-3-[[(4R)-2-oxo-4-thiazolidinyl]carbonyl]- 1-imidazolidinyl]phenyl]amino]-4-pyhmidinyl]amino]propyl]-2-oxo-4- thiazolidinecarboxamide.
11.A compound of following structure
N-(3-((4-((3-(aminomethyl)phenyl)amino)-5-bromo-2- pyrimidinyl)amino)phenyl)-1-pyrrolidine-carboxamide,
4-[[5-bromo-4-[[2-(1 H-imidazol-5-yl)ethyl]amino]-2-pyrimidinyl]amino]- 1 - naphthaleneacetic acid,
5-[[5-bromo-4-[[2-(1 H-imidazol-5-yl)ethyl]amino]-2-pyrimidinyl]amino]-1 H- indole-2-carboxylic acid, ethyl ester, 5-bromo-N4-[2-(1 H-imidazol-5-yl)ethyl]-N2-(2-methyl-6-quinolinyl)-2,4- pyrimidinediamine,
4-((5-bromo-4-((2-(7H-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzamide,
4-((4-((2-( H-imidazol-4-yl)ethyl)amino)-5-iodo-2-pyrimidinyl)amino)- benzenesulfonamide,
3-((5-bromo-4-((2-( /- -imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzamide,
3-((5-bromo-4-((2-( /-/-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide, 5-((5-bromo-4-((2-( /-/-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)-1 ,3- dihydro-2H-benzimidazol-2-one,
3-((5-bromo-4-((2-(f -/-imidazol-4-yl)ethyl)amino)-2-pyhmidinyl)amino)- benzoic acid methyl ester, 3-amino-5-((5-bromo-4-((2-(7H-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)- benzoic acid methyl ester,
/V-((3-((5-bromo-4-((2-(7tf-imidazol-4-yl)ethyl)amino)-2- pyrimidinyl)amino)phenyl)methyl)-methanesulfonamide, 4-((5-bromo-4-((2-(^H-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzoic acid methyl ester,
3-((5-bromo-4-((2-(' H-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- phenol,
5-((5-bromo-4-((2-(W-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)-' H- isoindole-1 ,3(2H)-dione,
5-bromo-/V4-(2-(W-imidazol-4-yl)ethyl)-Λ/2-(3-methylphenyl)-2,4- pyrimidinediamine,
/V-(3-((5-bromo-4-((2-(7H-imidazol-4-yl)ethyl)amino)-2- pyhmidinyl)amino)phenyl)-methanesulfonamide, 4-((4-((2-( 7H-imidazol-4-yl)ethyl)amino)-5-methyl-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((4-((2-(W-imidazol-4-yl)ethyl)amino)-5-(trifluoromethyl)-2- pyrimidinyl)amino)-benzenesulfonamide,
4-((4-((3-aminopropyl)amino)-5-bromo-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((5-bromo-4-((3-(' /-/-imidazol-1-yl)propyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((5-bromo-4-((2-(1-pyrrolidinyl)ethyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide, 4-((4-((4-aminobutyl)amino)-5-bromo-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyrimidinyl)amino)- butanoic acid,
4-((4-((3-((aminocarbonyl)amino)propyl)amino)-5-bromo-2- pyrimidinyl)amino)-benzenesulfonamide,
4-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyrimidinyl)amino)- butanoic acid ethyl ester,
4-((5-bromo-4-((4-(methylamino)butyl)amino)-2-pyhmidinyl)amino)- benzenesulfonamide,
4-((5-bromo-4-((2-(7H-imidazol-1-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((5-ethyl-4-((2-( H-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((4-((2-(7H-imidazol-4-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide,
4-((5-bromo-4-((2-(2-pyridinyl)ethyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide, 4-((5-bromo-4-((2-( -/-indol-3-yl)ethyl)amino)-2-pyrimidinyl)amino)- benzenesulfonamide,
2-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyhmidinyl)amino)- acetamide,
Λ/-(2-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4- pyrimidinyl)amino)ethyl)-acetamide,
3-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyrimidinyl)amino)- propanamide,
Λ/-(4-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4- pyrimidinyl)amino)butyl)-acetamide, Λ/-(3-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4- pyrimidinyl)amino)propyl)-acetamide,
Λ/-(3-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4- pyrimidinyl)amino)propyl)-2-furancarboxamide,
Λ/-(3-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4- pyrimidinyl)amino)propyl)-1H-pyrrole-2-carboxamide,
4-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4-pyhmidinyl)amino)- butanamide,
Λ/-(3-((2-((4-(aminosulfonyl)phenyl)amino)-5-bromo-4- pyrimidinyl)amino)propyl)-2-thiophenecarboxamide, 4-((4-(4-(aminomethyl)-1-pipehdinyl)-5-bromo-2-pyrimidinyl)amino)- benzenesulfonamide,
4-(5-Brom-4-prop-2-ynylamino-pyrimidin-2-ylamino)-phenyl]-N,N- dimethylaminosulfonylamin, 1 -Methyl-1 H-imidazol-4-sulfonsaure [4-(5-brom-4-prop-2-ynylamino- pyrimidin-2-ylamino)-phenyl]-amid,
3-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-benzoic acid ethyl ester,
4-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-benzoic acid ethyl ester, 2-(5-Bromo-4-prop-2-ynyloxy-pyhmidine-2-ylamino)-benzoic acid ethyl ester,
2-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-phenol,
4-(5-Bromo-4-prop-2-ynyloxy-pyrimidine-2-ylamino)-benzoic acid methyl ester,
3-(5-Nitro-4-prop-2-ynylamino-pyrimidine-2-ylamino)-phenol, 2-(5-Nitro-4-prop-2-ynylamino-pyrimidine-2-ylamino)-benzoic acid ethyl ester,
3-(5-Nitro-4-prop-2-ynylamino-pyhmidine-2-ylamino)-benzoic acid ethyl ester,
4-(5-Nitro-4-prop-2-ynylamino-pyrimidine-2-ylamino)-benzoic acid ethyl ester,
4-(5-Nitro-4-prop-2-ynylamino-pyrimidine-2-ylamino)-phenol,
Methyl 3-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]-5-[(2- hydroxyethyl)amino]benzoate,
Methyl 3-amino-5-[[5-bromo-4-(prop-2-ynyloxy)pyrimidin-2-yl]amino]benzoate or
3-[Bis-(2-hydroxy-ethyl)-amino]-5-(5-bromo-4-prop-2-ynyloxy-pyrimidine-2- ylamino)-benzoic acid methyl ester.
12. Pharmaceutical composition comprising as an active ingredient at least one compound of general formula (I) according to any one of claims 1 to 10 or compounds according to claim 11 in an therapeutically effective amount for the prevention or treatment of a disorder caused by, associated with or accompanied by disruptions of cell proliferation and/or angiogenesis together with an pharmaceutically acceptable carrier, diluent or excipient.
13. Use of a compound of general formula (I) according to claim 1 or 10 or compounds according to claim 11 for the manufacture of a medicament for the prevention or treatment of a disorder caused by, associated with or accompanied by any abnormal kinase activity selected from Chk, Akt, Pdk, Cdk and/or VEGF-R activity as well as combinations thereof.
14. The use of a compound of general formula (I) according to any one of claims 1 to 5, wherein the kinase is selected from PDK1 , Akt1 , Akt2 and/or Akt3.
15. The use of a compound of general formula (I) according to claim 13, wherein the kinase is selected from PDK1 , Akt1 , Akt2 and/or Akt3 in combination with
VEGF-R.
16. The use of a compound of general formula (I) according to any one of claims 1 and 6 to 8, wherein the kinase is selected from Chk1 and/or Chk2.
17. The use according to any one of claims 13 to 16, wherein the disorder is selected from cancer, angiofribroma, arthritis, eye diseases, auto-immune diseases, chemotherapy agent-induced alopecia and mucositis, Crohn- disease, endometriosis, fibrotic diseases, hemangioma, cardiovaskular diseases, infectious diseases, nephrological diseases, chronic und acute neurodegenerative diseases, like disruptions of nerval tissue, viral infections, to prevent restenosis of vessels, for preventing the formation of scars, preventing or treating keratoma seniles and contact dermatitis.
18. The use according to claim 17, wherein cancer stands for solide tumours, tumour- or metastasis growth, Kaposis
Sarkom, Hodgkin's disease and/or leukemia, arthritis stands for rheumatoid arthritis, eyes diseases stand for diabetic retinopathy, neovaskular glaukoma, auto-immune diseases stand for psoriasis, alopecia and/or multiple sklerosis, fibrotic diseases stand for cirrhosis of the liver, mesangial cell proliferative diseases, arteriosklerosis, infectiouse diseases stand for diseases that are caused by unicellular parasites, cardiovascular diseases stand for stenosis, like stent induced restenosis, arteriosklerosis and restenosis, nephrological diseases stand for glomerulonephritis, diabetic nephropaty, malignant nephrosklerosis, thrombic mikroangiopathis syndrome, transplant rejections and glomerulopathy, chronic neurodegenerative diseases stand for Huntington's disease, amyotrophic lateralsklerosis, Parkinsons disease, AIDS, dementia und Alzheimer's disease, acute neurodegenerative diseases stand for ischemias of the brain and neurotraumas, and viral infections stand for cytomegalic infections, herpes, hepatitis B or C and HIV.
19. A method of treating a mammal having a disease-state alleviated by the inhibition of Akt, Pdk, chk and/or VEGF-R activity, wherein the method comprises administering to a mammal a therapeutically effective amount of a compound of general formula (I) according to any one of claims 1 to 10 or the compounds of claim 11.
20. The method of claim 19 wherein the mammal is a human.
21. The method of claim 19 or 20, wherein the disease-state is cancer, angiofribroma, arthritis, eye diseases, auto-immune diseases, chemotherapy agent-induced alopecia and mucositis, Crohn's disease, endometriosis, fibrotic diseases, hemangioma, cardiovaskular diseases, infectious diseases, nephrological diseases, chronic und acute neurodegenerative diseases, like disruptions of nerval tissue, viral infections, prevention of restenosis of vessels, prevention the formation of scars, prevention or treatment of keratoma seniles or contact dermatitis.
22. The method of claim 21 , wherein cancer stands for solide tumours, tumour- or metastasis growth, Kaposis Sarkom, Hodgkin's disease and/or leukemia, arthritis stands for rheumatoid arthritis, eyes diseases stand for diabetic retinopathy, neovaskular glaukoma, auto-immune diseases stand for psoriasis, alopecia and/or multiple sklerosis, fibrotic diseases stand for cirrhosis of the liver, mesangial cell proliferative diseases, arteriosklerosis, infectiouse diseases stand for diseases that are caused by unicellular parasites, cardiovascular diseases stand for stenosis, like stent induced restenosis, arteriosklerosis and restenosis, nephrological diseases stand for glomerulonephritis, diabetic nephropaty, malignant nephrosklerosis, thrombic mikroangiopathis syndrome, transplant rejections and glomerulopathy, chronic neurodegenerative diseases stand for Huntington's disease, amyotrophic lateralsklerosis, Parkinsons disease, AIDS, dementia und
Alzheimer's disease, acute neurodegenerative diseases stand for ischemias of the brain and neurotraumas, and viral infections stand for cytomegalic infections, herpes, hepatitis B or C and HIV.
PCT/EP2003/013443 2002-11-28 2003-11-28 Chk-, pdk- and akt-inhibitory pyrimidines, their production and use as pharmaceutical agents WO2004048343A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
NZ539823A NZ539823A (en) 2002-11-28 2003-11-28 Chk-, Pdk- and Akt-inhibitory pyrimidines, their production and use as pharmaceutical agents
YUP-2005/0363A RS20050363A (en) 2002-11-28 2003-11-28 Chk-,pdk-and akt-inhibitory pyrimidines,their production an use as pharmaceutical agents
JP2004554522A JP2006508997A (en) 2002-11-28 2003-11-28 Chk-, Pdk- and Akt-inhibiting pyrimidines, their preparation and use as pharmaceuticals
EP03780086A EP1565446A1 (en) 2002-11-28 2003-11-28 Chk-, pdk- and akt-inhibitory pyrimidines, their production and use as pharmaceutical agents
AU2003288198A AU2003288198A1 (en) 2002-11-28 2003-11-28 CHK-,PDK- and AKT-inhibitory pyrimidines, their production and use as pharmaceutical agents
BR0316680-5A BR0316680A (en) 2002-11-28 2003-11-28 Chk, pdk and akt inhibitor pyrimidines, their production and use as pharmaceutical agents
CA002502970A CA2502970A1 (en) 2002-11-28 2003-11-28 Chk-, pdk- and akt-inhibitory pyrimidines, their production and use as pharmaceutical agents
MXPA05005547A MXPA05005547A (en) 2002-11-28 2003-11-28 Chk-, pdk- and akt-inhibitory pyrimidines, their production and use as pharmaceutical agents.
EA200500721A EA200500721A1 (en) 2002-11-28 2003-11-28 Pyrimidines inhibiting CHK, PDK and ACT, THEIR RECEIVING AND USING AS MEDICINES
IL168102A IL168102A0 (en) 2002-11-28 2005-04-17 Chk-, pdk-and akt-inhibitory pyrimidines, their production and use as pharmaceutical agents
IS7881A IS7881A (en) 2002-11-28 2005-06-02 CHK-, PDK- and AKT-inhibiting pyrimidines, their production and use as pharmaceuticals
NO20053144A NO20053144L (en) 2002-11-28 2005-06-27 Chk, Pdk, and Akt inhibitor pyrimidines, their preparation and use as famasoytic agents.
HR20050601A HRP20050601A2 (en) 2002-11-28 2005-06-28 Chk-, pdk-, and akt-inhibitory pyrimidines, their production and use as pharmarmaceutical agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP02026607 2002-11-28
EP02026607.8 2002-11-28

Publications (1)

Publication Number Publication Date
WO2004048343A1 true WO2004048343A1 (en) 2004-06-10

Family

ID=32338009

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2003/013443 WO2004048343A1 (en) 2002-11-28 2003-11-28 Chk-, pdk- and akt-inhibitory pyrimidines, their production and use as pharmaceutical agents

Country Status (20)

Country Link
US (1) US7504410B2 (en)
EP (1) EP1565446A1 (en)
JP (1) JP2006508997A (en)
KR (1) KR20050084027A (en)
CN (1) CN1717396A (en)
AU (1) AU2003288198A1 (en)
BR (1) BR0316680A (en)
CA (1) CA2502970A1 (en)
EA (1) EA200500721A1 (en)
EC (1) ECSP055899A (en)
HR (1) HRP20050601A2 (en)
IL (1) IL168102A0 (en)
IS (1) IS7881A (en)
MX (1) MXPA05005547A (en)
NO (1) NO20053144L (en)
NZ (1) NZ539823A (en)
PL (1) PL377795A1 (en)
RS (1) RS20050363A (en)
WO (1) WO2004048343A1 (en)
ZA (1) ZA200505184B (en)

Cited By (84)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005040116A2 (en) * 2003-10-24 2005-05-06 Schering Aktiengesellschaft Indolinone derivatives and their use in treating disease-states such as cancer
EP1624873A2 (en) * 2003-04-28 2006-02-15 AB Science Use of tyrosine kinase inhibitors for treating cerebral ischemia
JP2006515298A (en) * 2002-12-20 2006-05-25 ファイザー・プロダクツ・インク Pyrimidine derivatives for treating abnormal cell proliferation
US7060827B2 (en) 2002-02-01 2006-06-13 Rigel Pharmaceuticals, Inc. Intermediates useful for making 2,4-pyrimidinediamine compounds
WO2006076706A1 (en) * 2005-01-14 2006-07-20 Millennium Pharmaceuticals, Inc. Cinnamide and hydrocinnamide derivatives with raf-kinase inhibitory activity
WO2006105262A1 (en) 2005-03-29 2006-10-05 Icos Corporation HETEROARYL UREA DERIVATIVES USEFUL FOR INHIBITING CHKl
US7122542B2 (en) 2003-07-30 2006-10-17 Rigel Pharmaceuticals, Inc. Methods of treating or preventing autoimmune diseases with 2,4-pyrimidinediamine compounds
US7157455B2 (en) 2003-02-10 2007-01-02 Hoffmann-La Roche Inc. 4-Aminopyrimidine-5-one derivatives
WO2007030362A1 (en) * 2005-09-07 2007-03-15 Laboratoires Serono Sa. Ikk inhibitors for the treatment of endometriosis
WO2007047146A2 (en) * 2005-10-11 2007-04-26 Intermune, Inc. Inhibitors of viral replication
WO2007146981A2 (en) * 2006-06-15 2007-12-21 Boehringer Ingelheim International Gmbh 2-anilino-4-(heterocyclic)amino-pyrimidines as inhibitors of protein kinase c-alpha
EP1890703A2 (en) * 2005-06-14 2008-02-27 Taigen Biotechnology Pyrimidine compounds
JP2008514680A (en) * 2004-09-30 2008-05-08 テイボテク・フアーマシユーチカルズ・リミテツド HIV inhibitory 5-carbocyclic or heterocyclic substituted pyrimidines
WO2008129380A1 (en) 2007-04-18 2008-10-30 Pfizer Products Inc. Sulfonyl amide derivatives for the treatment of abnormal cell growth
WO2009053694A1 (en) * 2007-10-24 2009-04-30 Cancer Research Technology Limited Therapeutic oxy-phenyl-aryl compounds and their use
JP2009520740A (en) * 2005-12-22 2009-05-28 バイエル・シエーリング・ファーマ アクチエンゲゼルシャフト Sulfoximine substituted pyrimidines, their preparation and use as pharmaceuticals
WO2009071535A1 (en) * 2007-12-03 2009-06-11 Boehringer Ingelheim International Gmbh Diaminopyridines for the treatment of diseases which are characterised by excessive or anomal cell proliferation
WO2009122180A1 (en) * 2008-04-02 2009-10-08 Medical Research Council Pyrimidine derivatives capable of inhibiting one or more kinases
JP2009540012A (en) * 2006-06-15 2009-11-19 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング 2-anilino-4-aminoalkyleneaminopyrimidine
EP2136635A1 (en) * 2007-03-20 2009-12-30 Smithkline Beecham Corporation Chemical compounds
WO2010024430A1 (en) * 2008-09-01 2010-03-04 アステラス製薬株式会社 2,4-diaminopyrimidine compound
EP2161259A1 (en) * 2008-09-03 2010-03-10 Bayer CropScience AG 4-Haloalkyl substituted Diaminopyrimidine
WO2010025831A1 (en) * 2008-09-03 2010-03-11 Bayer Cropscience Ag Heterocyclically substituted anilinopyrimidines as fungicides
WO2010027448A1 (en) * 2008-09-02 2010-03-11 Actimis Pharmaceuticals Inc. Isotopically enriched pyrimidin-5-yl acetic acid derivatives as crth2 antagonists
WO2010025833A1 (en) * 2008-09-03 2010-03-11 Bayer Cropscience Ag Alkoxy-substituted and alkylthio-substituted anilinopyrimidines
US7705009B2 (en) 2005-11-22 2010-04-27 Hoffman-La Roche Inc. 4-aminopyrimidine-5-thione derivatives
EP2179992A1 (en) 2008-10-21 2010-04-28 Bayer Schering Pharma Aktiengesellschaft Sulfon substituted aniline pyrimidine derivatives as CDK inhibitors, their manufacture and use as medicine
EP2179991A1 (en) 2008-10-21 2010-04-28 Bayer Schering Pharma Aktiengesellschaft Sulfoximine substituted aniline pyrimidine derivatives as CDK inhibitors, their manufacture and use as medicine
WO2010058030A1 (en) * 2008-11-24 2010-05-27 Boehringer Ingelheim International Gmbh New compounds
DE102009001438A1 (en) 2009-03-10 2010-09-16 Bayer Schering Pharma Aktiengesellschaft New carbonylamino-substituted anilino-pyrimidine compounds are tyrosine kinase-2 inhibitors, useful for treating diseases associated with inflammatory conditions e.g. bronchitis, rheumatoid arthritis, psoriasis and Guillain Barre syndrome
US7812029B1 (en) 2002-07-29 2010-10-12 Rigel Pharmaceuticals, Inc. Methods of treating or preventing autoimmune diseases with 2,4-pyrimidinediamine compounds
DE102009015070A1 (en) 2009-03-30 2010-10-14 Bayer Schering Pharma Aktiengesellschaft New phenyl-pyrimidin-2-yl-amine compounds are tyrosine kinase 2 inhibitors useful for treating e.g. rheumatoid arthritis, Crohn's disease, asthma, multiple sclerosis, adult respiratory distress syndrome, allergic alveolitis and uveitis
US7851480B2 (en) 2004-11-24 2010-12-14 Rigel Pharmaceuticals, Inc. Spiro 2,4-pyrimidinediamine compounds and their uses
WO2011018517A1 (en) 2009-08-14 2011-02-17 Boehringer Ingelheim International Gmbh Regioselective preparation of 2-amino-5-trifluoromethylpyrimidine derivatives
WO2011018518A1 (en) 2009-08-14 2011-02-17 Boehringer Ingelheim International Gmbh Regioselective preparation of 2 -amino-5-trifluoromethylpyrimidine derivatives
US7989448B2 (en) 2005-01-19 2011-08-02 Rigel Pharmaceuticals, Inc. Prodrugs of 2,4-pyrimidinediamine compounds and their uses
US8088771B2 (en) 2008-07-28 2012-01-03 Gilead Sciences, Inc. Cycloalkylidene and heterocycloalkylidene inhibitor compounds
US8124764B2 (en) 2008-07-14 2012-02-28 Gilead Sciences, Inc. Fused heterocyclyc inhibitor compounds
US8134000B2 (en) 2008-07-14 2012-03-13 Gilead Sciences, Inc. Imidazolyl pyrimidine inhibitor compounds
US8258316B2 (en) 2009-06-08 2012-09-04 Gilead Sciences, Inc. Alkanoylamino benzamide aniline HDAC inhibitor compounds
US8283357B2 (en) 2009-06-08 2012-10-09 Gilead Sciences, Inc. Cycloalkylcarbamate benzamide aniline HDAC inhibitor compounds
EP2519664A1 (en) * 2009-12-30 2012-11-07 Avila Therapeutics, Inc. Ligand-directed covalent modification of protein
US20120329771A1 (en) * 2010-11-18 2012-12-27 Boehringer Ingelheim International Gmbh Benzylic oxindole pyrimidines
US8344018B2 (en) 2008-07-14 2013-01-01 Gilead Sciences, Inc. Oxindolyl inhibitor compounds
US8354420B2 (en) 2010-06-04 2013-01-15 Genentech, Inc. Aminopyrimidine derivatives as LRRK2 inhibitors
US8377943B2 (en) 2008-11-14 2013-02-19 Boehringer Ingelheim International Gmbh 2,4-diaminopyrimidine derivates as PTK2-inhibitors for the treatment of abnormal cell growth
WO2013041537A1 (en) 2011-09-23 2013-03-28 Beiersdorf Ag Heterocyclocarbonylaminothiazoles, cosmetic or dermatological preparations containing said heterocyclocarbonylaminothiazoles, and use thereof to combat or prevent undesired pigmentation of the skin
WO2013157540A1 (en) * 2012-04-17 2013-10-24 富士フイルム株式会社 Nitrogen-containing heterocyclic compound or salt thereof
US8815882B2 (en) 2010-11-10 2014-08-26 Genentech, Inc. Pyrazole aminopyrimidine derivatives as LRRK2 modulators
US8846681B2 (en) 2006-07-06 2014-09-30 Array Biopharma, Inc. Pyrimidyl cyclopentanes as AKT protein kinase inhibitors
US8846689B2 (en) 2008-11-24 2014-09-30 Boehringer Ingelheim International Gmbh Substituted pyrimidines for the treatment of diseases such as cancer
US8901143B2 (en) 2007-04-16 2014-12-02 Hutchison Medipharma Enterprises Limited Pyrimidine derivatives
US9150548B2 (en) 2011-04-01 2015-10-06 Genentech, Inc. Combinations of AKT inhibitor compounds and vemurafenib, and methods of use
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
US9388185B2 (en) 2012-08-10 2016-07-12 Incyte Holdings Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9533984B2 (en) 2013-04-19 2017-01-03 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US9533954B2 (en) 2010-12-22 2017-01-03 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US9556426B2 (en) 2009-09-16 2017-01-31 Celgene Avilomics Research, Inc. Protein kinase conjugates and inhibitors
US9580423B2 (en) 2015-02-20 2017-02-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9611267B2 (en) 2012-06-13 2017-04-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US9682082B2 (en) 2011-04-01 2017-06-20 Genentech, Inc. Combinations of AKT and MEK inhibitor compounds, and methods of use
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
EP3177295A4 (en) * 2014-08-08 2018-06-13 Dana Farber Cancer Institute, Inc. Uses of salt-inducible kinase (sik) inhibitors
US10011571B2 (en) 2014-09-16 2018-07-03 Shenzhen Chipscreen Biosciences, Ltd. Preparation method for aromatic heterocyclic compound used as selective JAK3 and/or JAK1 kinase inhibitor and application of aromatic heterocyclic compound
US10233157B2 (en) 2012-03-13 2019-03-19 Dana-Farber Cancer Institute, Inc. SIK inhibitor for use in a method of treating an inflammatory and/or immune disorder
US10287268B2 (en) 2014-07-21 2019-05-14 Dana-Farber Cancer Institute, Inc. Imidazolyl kinase inhibitors and uses thereof
EP3030241A4 (en) * 2013-08-06 2019-06-19 H. Lee Moffitt Cancer Center And Research Institute, Inc. Inhibitors of ack1/tnk2 tyrosine kinase
US10457691B2 (en) 2014-07-21 2019-10-29 Dana-Farber Cancer Institute, Inc. Macrocyclic kinase inhibitors and uses thereof
WO2020023851A1 (en) * 2018-07-26 2020-01-30 Yale University Bifunctional substitued pyrimidines as modulators of fak proteolyse
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
WO2020231806A1 (en) * 2019-05-10 2020-11-19 Deciphera Pharmaceuticals, Llc Phenylaminopyrimidine amide autophagy inhibitors and methods of use thereof
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10954242B2 (en) 2016-07-05 2021-03-23 The Broad Institute, Inc. Bicyclic urea kinase inhibitors and uses thereof
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11241435B2 (en) 2016-09-16 2022-02-08 The General Hospital Corporation Uses of salt-inducible kinase (SIK) inhibitors for treating osteoporosis
US11285158B2 (en) 2017-02-28 2022-03-29 The General Hospital Corporation Uses of pyrimidopyrimidinones as SIK inhibitors
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11591329B2 (en) 2019-07-09 2023-02-28 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors

Families Citing this family (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1392662B1 (en) * 2001-05-29 2009-01-07 Bayer Schering Pharma Aktiengesellschaft Cdk inhibiting pyrimidines, production thereof and their use as medicaments
US7332521B2 (en) * 2003-09-25 2008-02-19 Wyeth Substituted indoles
JP2008523103A (en) * 2004-12-14 2008-07-03 バーテックス ファーマシューティカルズ インコーポレイテッド Pyrimidine inhibitor of ERK protein kinase and use thereof
US8716299B2 (en) * 2004-12-20 2014-05-06 University Of South Florida XIAP-targeted prostate cancer therapy
AP2008004488A0 (en) * 2005-12-21 2008-06-30 Pfizer Prod Inc Pyrimidine derivatives for the treatment of abnormal cell growth
US8426355B2 (en) * 2006-03-15 2013-04-23 Theralogics, Inc. Methods of treating muscular wasting diseases using NF-κB activation inhibitors
DE102006027156A1 (en) * 2006-06-08 2007-12-13 Bayer Schering Pharma Ag New sulfimide compounds are protein kinase inhibitors useful to treat e.g. cancer, Hodgkin's lymphoma, Kaposi's sarcoma, cardiovascular disease, Crohn's disease, endometriosis and hemangioma
CA2669982A1 (en) * 2006-11-17 2008-05-29 Schering Corporation Combination therapy for proliferative disorders
WO2009030890A1 (en) * 2007-09-03 2009-03-12 University Court Of The University Of Dundee Pyrimidine compounds for the treatment of cancer, septic shock and/or primary open angle glaucoma
PL2639226T3 (en) * 2007-09-17 2017-02-28 Abbvie Bahamas Ltd. Anti-infective pyrimidines and uses thereof
US20110065776A1 (en) * 2009-09-17 2011-03-17 Keng-Hsin Lan Method for Treating Hepatitis C Infection
BR112012029647A2 (en) 2010-05-21 2016-08-02 Chemilia Ab new pyrimidine derivatives
JPWO2012005299A1 (en) * 2010-07-07 2013-09-05 日本新薬株式会社 ROS tyrosine kinase inhibitor
CA2830129C (en) 2011-03-24 2016-07-19 Chemilia Ab Novel pyrimidine derivatives
JP6180426B2 (en) * 2011-11-29 2017-08-16 ジェネンテック, インコーポレイテッド 2- (Phenyl or pyrid-3-yl) aminopyrimidine derivatives as kinase LRRK2 modulators for the treatment of Parkinson's disease
AR089182A1 (en) * 2011-11-29 2014-08-06 Hoffmann La Roche AMINOPIRIMIDINE DERIVATIVES AS MODULATORS OF LRRK2
CN102746337B (en) * 2012-06-21 2014-12-17 成都苑东药业有限公司 2,4-pyrimidinediamine compound and preparation method thereof
WO2014022185A2 (en) * 2012-08-03 2014-02-06 Albert Einstein College Of Medicine Of Yeshiva University Method to treat or prevent herpesvirus infections
US10130632B2 (en) * 2012-11-27 2018-11-20 Beth Israel Deaconess Medical Center, Inc. Methods for treating renal disease
CN103539744B (en) * 2013-10-17 2016-03-23 北京颖泰嘉和生物科技股份有限公司 One prepares the method for 3-(3,5-dichlorophenyl)-2,4-imidazolidimediones
EP3099684B8 (en) * 2014-01-29 2018-07-04 UCB Biopharma SPRL Heteroaryl amides as inhibitors of protein aggregation
GB201402277D0 (en) * 2014-02-10 2014-03-26 Sentinel Oncology Ltd Pharmaceutical compounds
JP6431294B2 (en) * 2014-06-16 2018-11-28 東ソー・ファインケム株式会社 5- (Trifluoromethyl) pyrimidine derivative and process for producing the same
CN105481778B (en) * 2014-09-16 2019-06-04 深圳微芯生物科技股份有限公司 Pyrimidine derivatives, preparation method and its application
CA2962914C (en) * 2014-10-13 2018-10-23 Yuhan Corporation Compounds and compositions for modulating egfr mutant kinase activities
JP6479486B2 (en) * 2015-01-16 2019-03-06 東ソー・ファインケム株式会社 4-alkoxy-5- (trifluoromethyl) pyrimidine derivative and method for producing the same
MX2017016619A (en) * 2015-07-09 2018-05-15 Merck Patent Gmbh Pyrimidine derivatives as btk inhibitors and uses thereof.
US20190290656A1 (en) 2015-12-30 2019-09-26 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd Pt(iv) prodrugs
CN106083828A (en) * 2016-06-30 2016-11-09 浙江大学 Arylamine pyrimidine derivatives containing ethylene imine ring side chain and preparation and application
KR101876514B1 (en) * 2016-11-08 2018-07-10 한국화학연구원 Novel pyrimidine compounds, preparation method thereof, and pharmaceutical composition for use in preventing or treating cancer and inflammation disease containing the same as an active ingredient
CN109928935B (en) * 2017-12-19 2023-03-10 沈阳化工研究院有限公司 Anilino pyrimidine compound and medical application thereof
HRP20231310T1 (en) 2019-05-10 2024-02-02 Deciphera Pharmaceuticals, Llc Heteroarylaminopyrimidine amide autophagy inhibitors and methods of use thereof
CA3143489A1 (en) 2019-06-17 2020-12-24 Deciphera Pharmaceuticals, Llc Aminopyrimidine amide autophagy inhibitors and methods of use thereof
KR20240017731A (en) * 2022-08-01 2024-02-08 동아대학교 산학협력단 Use of PDH inhibitors for treatment of neurodegenerative disease

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001072717A1 (en) * 2000-03-28 2001-10-04 Astrazeneca Ab 4-amino-5-cyano-2-anilino-pyrimidine derivatives and their use as inhibitors of cell-cycle kinases
WO2002004429A1 (en) * 2000-07-11 2002-01-17 Astrazeneca Ab Pyrimidine derivatives

Family Cites Families (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3055900A (en) * 1957-12-06 1962-09-25 Ciba Geigy Corp Pyrimido pyrimidines and process of making same
ES477520A1 (en) * 1978-02-25 1979-06-01 Thomae Gmbh Dr K Penicillins, their salts, process for their preparation and pharmaceutical compositions containing them.
JPS60100559A (en) * 1983-11-05 1985-06-04 Morishita Seiyaku Kk 2-anilino-1,6-dihydro-6-oxo-5-pyrimidinecarboxylic acid derivative, its preparation, and antiallergic containing it
JPS6110563A (en) * 1984-06-25 1986-01-18 Hokuriku Seiyaku Co Ltd Pyrimidylaminophenylpropionic acid derivative
JPH0625161B2 (en) * 1984-10-09 1994-04-06 森下ルセル株式会社 Process for producing 2-anilino-1,6-dihydro-6-oxo-5-pyrimidinecarboxylic acid derivative
JPS61112059A (en) * 1984-11-06 1986-05-30 Morishita Seiyaku Kk 2-anilino-1,6-dihydro-6-oxo-5-pyrimidinecarbonitrile derivative
GB8506318D0 (en) * 1985-03-12 1985-04-11 Smith Kline French Lab Chemical compounds
JPH0629263B2 (en) * 1985-10-30 1994-04-20 クミアイ化学工業株式会社 Pyrimidine derivatives and agricultural and horticultural fungicides
JPH03127790A (en) * 1989-10-11 1991-05-30 Morishita Pharmaceut Co Ltd N-(1h-tetrazol-5-yl)-2-anilino-5-pyrimidinecarboxamides and synthetic intermediate therefor
DE4029650A1 (en) * 1990-09-19 1992-03-26 Hoechst Ag New 2-aryl:amino-pyrimidine derivs. - contg. alkynyl gp., useful as fungicides
KR960704855A (en) * 1993-10-12 1996-10-09 돈 엠. 커. 1N-Alkyl-N-arylpyrimidinamines and derivatives thereof
US5516905A (en) * 1994-08-30 1996-05-14 University Of Massachusetts Medical Center Antibiotic compounds and methods to treat gram-positive bacterial and mycoplasmal infections
IL115256A0 (en) * 1994-11-14 1995-12-31 Warner Lambert Co 6-Aryl pyrido (2,3-d) pyrimidines and naphthyridines and their use
IL117923A (en) * 1995-05-03 2000-06-01 Warner Lambert Co Anti-cancer pharmaceutical compositions containing polysubstituted pyrido¬2,3-d¾pyrimidine derivatives and certain such novel compounds
US5620981A (en) * 1995-05-03 1997-04-15 Warner-Lambert Company Pyrido [2,3-D]pyrimidines for inhibiting protein tyrosine kinase mediated cellular proliferation
CA2271157A1 (en) * 1997-02-05 1998-08-06 Warner-Lambert Company Pyrido[2,3-d]pyrimidines and 4-aminopyrimidines as inhibitors of cellular proliferation
AU7237398A (en) * 1997-05-15 1998-12-08 Mitsubishi Chemical Corporation Anilinopyrimidinone derivatives, processes for producing the same, and insecticidal/acaricidal agents containing the same as active ingredient
KR100643419B1 (en) * 1998-03-27 2006-11-10 얀센 파마슈티카 엔.브이. ??? inhibiting pyrimidine derivatives
SI0945443T1 (en) * 1998-03-27 2003-08-31 Janssen Pharmaceutica N.V. HIV inhibiting pyrimidine derivatives
CZ301367B6 (en) * 1998-11-10 2010-02-03 Janssen Pharmaceutica N. V. Pyrimidine derivative, process and intermediates for its preparation and medicament based thereon and intended for treating HIV
CN1243002C (en) * 1998-11-17 2006-02-22 组合化学工业株式会社 Pyrimidinylbenzimidazole and triazinylbenzimidazole derivatives and agricultural/horticultural bactericides
HUP0202017A3 (en) * 1999-03-26 2003-03-28 Astrazeneca Ab Novel compounds
PL353269A1 (en) * 1999-09-15 2003-11-03 Warner-Lambert Company Pteridinones as kinase inhibitors
NZ517025A (en) * 1999-09-24 2003-07-25 Janssen Pharmaceutica Nv Antiviral compositions
EA200200643A1 (en) * 2000-01-25 2002-12-26 Уорнер-Ламберт Компани PYRIDO [2,3-d] PYRIMIDIN-2,7-DIAMINE KINAZ INHIBITORS
HUP0203803A3 (en) * 2000-01-27 2004-09-28 Warner Lambert Co Pyridopyrimidinone derivatives for treatment of neurodegenerative disease
DK1282607T3 (en) * 2000-05-08 2016-02-01 Janssen Pharmaceutica Nv Prodrugs of HIV replication inhibiting pyrimidines
EP1392662B1 (en) * 2001-05-29 2009-01-07 Bayer Schering Pharma Aktiengesellschaft Cdk inhibiting pyrimidines, production thereof and their use as medicaments
JP2005500294A (en) * 2001-06-19 2005-01-06 ブリストル−マイヤーズ スクイブ カンパニー Pyrimidine inhibitors for phosphodiesterase 7
EP1406875B1 (en) * 2001-06-26 2013-07-31 Bristol-Myers Squibb Company N-heterocyclic inhibitors of tnf-alpha expression
JO3429B1 (en) * 2001-08-13 2019-10-20 Janssen Pharmaceutica Nv Hiv inhibiting pyrimidines derivatives
EP1453516A2 (en) * 2001-10-17 2004-09-08 Boehringer Ingelheim Pharma GmbH & Co.KG Novel tri-substituted pyrimidines, method for production and use thereof as medicament
EP2332924A1 (en) * 2001-10-17 2011-06-15 Boehringer Ingelheim Pharma GmbH & Co. KG Pyrimidine derivates, medicaments comprising them, their use and process of their preparation
EP1448556A1 (en) * 2001-11-01 2004-08-25 Janssen Pharmaceutica N.V. Heteroaryl amines as glycogen synthase kinase 3beta inhibitors (gsk3 inhibitors)
PT1442024E (en) * 2001-11-01 2008-06-12 Janssen Pharmaceutica Nv Aminobenzamide derivatives as glycogen synthase kinase 3$g(b) inhibitors
TWI329105B (en) * 2002-02-01 2010-08-21 Rigel Pharmaceuticals Inc 2,4-pyrimidinediamine compounds and their uses
AU2003265336B8 (en) * 2002-07-29 2009-04-23 Rigel Pharmaceuticals, Inc. Methods of treating or preventing autoimmune diseases with 2,4-pyrimidinediamine compounds
EP1529032B1 (en) * 2002-08-09 2013-04-24 Janssen Pharmaceutica NV Processes for the preparation of 4- 4- 4-(2 -cyanoethenyl)-2,6-dimethylphenyl )amino) -2-pyrimidinyl)amino benzonitrile

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001072717A1 (en) * 2000-03-28 2001-10-04 Astrazeneca Ab 4-amino-5-cyano-2-anilino-pyrimidine derivatives and their use as inhibitors of cell-cycle kinases
WO2002004429A1 (en) * 2000-07-11 2002-01-17 Astrazeneca Ab Pyrimidine derivatives

Cited By (169)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9346765B2 (en) 2002-02-01 2016-05-24 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and their uses
US10682350B2 (en) 2002-02-01 2020-06-16 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and their uses
US7655797B2 (en) 2002-02-01 2010-02-02 Rigel Pharmaceuticals, Inc. Intermediates for making 2,4-pyrimidinediamine compounds
US7803939B2 (en) 2002-02-01 2010-09-28 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and their uses
US7060827B2 (en) 2002-02-01 2006-06-13 Rigel Pharmaceuticals, Inc. Intermediates useful for making 2,4-pyrimidinediamine compounds
US7820819B2 (en) 2002-02-01 2010-10-26 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and their uses
US10709703B2 (en) 2002-02-01 2020-07-14 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and their uses
US9913842B2 (en) 2002-02-01 2018-03-13 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and their uses
US7906644B2 (en) 2002-02-01 2011-03-15 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and their uses
US9416112B2 (en) 2002-02-01 2016-08-16 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and their uses
US8835430B2 (en) 2002-02-01 2014-09-16 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and their uses
US8334296B2 (en) 2002-02-01 2012-12-18 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and their uses
US9018204B1 (en) 2002-02-01 2015-04-28 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and their uses
US7825116B2 (en) 2002-07-29 2010-11-02 Rigel Pharmaceuticals, Inc. N2, N4-bis-aryl-5-fluoro-2,4-pyrimidinediamines
US8557806B2 (en) 2002-07-29 2013-10-15 Rigel Pharmaceuticals, Inc. Methods of treating or preventing autoimmune diseases with 2,4-pyrimidinediamine compounds
US7812029B1 (en) 2002-07-29 2010-10-12 Rigel Pharmaceuticals, Inc. Methods of treating or preventing autoimmune diseases with 2,4-pyrimidinediamine compounds
JP2006515298A (en) * 2002-12-20 2006-05-25 ファイザー・プロダクツ・インク Pyrimidine derivatives for treating abnormal cell proliferation
US7157455B2 (en) 2003-02-10 2007-01-02 Hoffmann-La Roche Inc. 4-Aminopyrimidine-5-one derivatives
US7615634B2 (en) 2003-02-10 2009-11-10 Hoffmann-La Roche Inc. 4-aminopyrimidine-5-one derivatives
EP1624873A2 (en) * 2003-04-28 2006-02-15 AB Science Use of tyrosine kinase inhibitors for treating cerebral ischemia
US9751893B2 (en) 2003-07-30 2017-09-05 Rigel Pharmaceuticals, Inc. Methods of treating or preventing autoimmune diseases with 2,4-pyrimidinediamine compounds
US7122542B2 (en) 2003-07-30 2006-10-17 Rigel Pharmaceuticals, Inc. Methods of treating or preventing autoimmune diseases with 2,4-pyrimidinediamine compounds
WO2005040116A2 (en) * 2003-10-24 2005-05-06 Schering Aktiengesellschaft Indolinone derivatives and their use in treating disease-states such as cancer
US7105563B2 (en) 2003-10-24 2006-09-12 Schering Aktiengesellschaft Indolinone derivatives and their use in treating disease-states such as cancer
WO2005040116A3 (en) * 2003-10-24 2005-06-16 Schering Ag Indolinone derivatives and their use in treating disease-states such as cancer
JP2008514680A (en) * 2004-09-30 2008-05-08 テイボテク・フアーマシユーチカルズ・リミテツド HIV inhibitory 5-carbocyclic or heterocyclic substituted pyrimidines
US7851480B2 (en) 2004-11-24 2010-12-14 Rigel Pharmaceuticals, Inc. Spiro 2,4-pyrimidinediamine compounds and their uses
US7652041B2 (en) 2005-01-14 2010-01-26 Millennium Pharmaceuticals, Inc. Cinnamide and hydrocinnamide derivatives with kinase inhibitory activity
WO2006076706A1 (en) * 2005-01-14 2006-07-20 Millennium Pharmaceuticals, Inc. Cinnamide and hydrocinnamide derivatives with raf-kinase inhibitory activity
US8211888B2 (en) 2005-01-19 2012-07-03 Rigel Pharmaceuticals, Inc. Prodrugs of 2,4-pyrimidinediamine compounds and their uses
US8211889B2 (en) 2005-01-19 2012-07-03 Rigel Pharmaceuticals, Inc. Prodrugs of 2,4-pyrimidinediamine compounds and their uses
US8476263B2 (en) 2005-01-19 2013-07-02 Rigel Pharmaceuticals, Inc. Prodrugs of 2,4-pyrimidinediamine compounds and their uses
US8785437B2 (en) 2005-01-19 2014-07-22 Rigel Pharmaceuticals, Inc. Prodrugs of 2,4-pyrimidinediamine compounds and their uses
US9266912B2 (en) 2005-01-19 2016-02-23 Rigel Pharmaceuticals, Inc. Prodrugs of 2,4-pyrimidinediamine compounds and their uses
US9532998B2 (en) 2005-01-19 2017-01-03 Rigel Pharmaceuticals, Inc. Prodrugs of 2,4-pyrimidinediamine compounds and their uses
US7989448B2 (en) 2005-01-19 2011-08-02 Rigel Pharmaceuticals, Inc. Prodrugs of 2,4-pyrimidinediamine compounds and their uses
US10577381B2 (en) 2005-01-19 2020-03-03 Rigel Pharmaceuticals, Inc. Prodrugs of 2,4-pyrimidinediamine compounds and their uses
EP2330105A1 (en) 2005-03-29 2011-06-08 ICOS Corporation Heteroaryl urea derivatives useful for inhibiting CHK1
WO2006105262A1 (en) 2005-03-29 2006-10-05 Icos Corporation HETEROARYL UREA DERIVATIVES USEFUL FOR INHIBITING CHKl
EP1890703B1 (en) * 2005-06-14 2016-05-11 Taigen Biotechnology Pyrimidine compounds as chemokine receptors inhibitors
EP1890703A2 (en) * 2005-06-14 2008-02-27 Taigen Biotechnology Pyrimidine compounds
WO2007030362A1 (en) * 2005-09-07 2007-03-15 Laboratoires Serono Sa. Ikk inhibitors for the treatment of endometriosis
WO2007047146A3 (en) * 2005-10-11 2007-11-01 Intermune Inc Inhibitors of viral replication
WO2007047146A2 (en) * 2005-10-11 2007-04-26 Intermune, Inc. Inhibitors of viral replication
US7705009B2 (en) 2005-11-22 2010-04-27 Hoffman-La Roche Inc. 4-aminopyrimidine-5-thione derivatives
JP2009520740A (en) * 2005-12-22 2009-05-28 バイエル・シエーリング・ファーマ アクチエンゲゼルシャフト Sulfoximine substituted pyrimidines, their preparation and use as pharmaceuticals
JP2009540012A (en) * 2006-06-15 2009-11-19 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング 2-anilino-4-aminoalkyleneaminopyrimidine
WO2007146981A2 (en) * 2006-06-15 2007-12-21 Boehringer Ingelheim International Gmbh 2-anilino-4-(heterocyclic)amino-pyrimidines as inhibitors of protein kinase c-alpha
WO2007146981A3 (en) * 2006-06-15 2008-02-21 Boehringer Ingelheim Int 2-anilino-4-(heterocyclic)amino-pyrimidines as inhibitors of protein kinase c-alpha
US8354407B2 (en) 2006-06-15 2013-01-15 Boehringer Ingelheim International Gmbh 2-anilino-4-(heterocyclic)amino-pyrimidines
US8158641B2 (en) 2006-06-15 2012-04-17 Boehringer Ingelheim International Gmbh 2-anilino-4-aminoalkyleneaminopyrimidines
JP2009540013A (en) * 2006-06-15 2009-11-19 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング 2-anilino-4- (heterocyclic) amino-pyrimidine
US8846681B2 (en) 2006-07-06 2014-09-30 Array Biopharma, Inc. Pyrimidyl cyclopentanes as AKT protein kinase inhibitors
EP2136635A4 (en) * 2007-03-20 2011-07-27 Glaxosmithkline Llc Chemical compounds
EP2136635A1 (en) * 2007-03-20 2009-12-30 Smithkline Beecham Corporation Chemical compounds
US8901143B2 (en) 2007-04-16 2014-12-02 Hutchison Medipharma Enterprises Limited Pyrimidine derivatives
KR101132880B1 (en) 2007-04-18 2012-06-28 화이자 프로덕츠 인크. Sulfonyl amide derivatives for the treatment of abnormal cell growth
US7928109B2 (en) 2007-04-18 2011-04-19 Pfizer Inc Sulfonyl amide derivatives for the treatment of abnormal cell growth
EA016679B1 (en) * 2007-04-18 2012-06-29 Пфайзер Продактс Инк. Sulfonyl amide derivatives for the treatment of abnormal cell growth
US8440822B2 (en) 2007-04-18 2013-05-14 Michael Joseph Luzzio Sulfonyl amide derivatives for the treatment of abnormal cell growth
US10450297B2 (en) 2007-04-18 2019-10-22 Pfizer, Inc. Sulfonyl amide derivatives for the treatment of abnormal cell growth
US8247411B2 (en) 2007-04-18 2012-08-21 Pfizer Inc Sulfonyl amide derivatives for the treatment of abnormal cell growth
WO2008129380A1 (en) 2007-04-18 2008-10-30 Pfizer Products Inc. Sulfonyl amide derivatives for the treatment of abnormal cell growth
WO2009053694A1 (en) * 2007-10-24 2009-04-30 Cancer Research Technology Limited Therapeutic oxy-phenyl-aryl compounds and their use
US8461147B2 (en) 2007-12-03 2013-06-11 Boehringer Ingelheim International Gmbh Diaminopyridines for the treatment of diseases which are characterised by excessive or anomal cell proliferation
JP2011505407A (en) * 2007-12-03 2011-02-24 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Diaminopyridine for treating diseases characterized by excessive or abnormal cell proliferation
WO2009071535A1 (en) * 2007-12-03 2009-06-11 Boehringer Ingelheim International Gmbh Diaminopyridines for the treatment of diseases which are characterised by excessive or anomal cell proliferation
WO2009122180A1 (en) * 2008-04-02 2009-10-08 Medical Research Council Pyrimidine derivatives capable of inhibiting one or more kinases
US8134000B2 (en) 2008-07-14 2012-03-13 Gilead Sciences, Inc. Imidazolyl pyrimidine inhibitor compounds
US8344018B2 (en) 2008-07-14 2013-01-01 Gilead Sciences, Inc. Oxindolyl inhibitor compounds
US8124764B2 (en) 2008-07-14 2012-02-28 Gilead Sciences, Inc. Fused heterocyclyc inhibitor compounds
US8088771B2 (en) 2008-07-28 2012-01-03 Gilead Sciences, Inc. Cycloalkylidene and heterocycloalkylidene inhibitor compounds
WO2010024430A1 (en) * 2008-09-01 2010-03-04 アステラス製薬株式会社 2,4-diaminopyrimidine compound
US8541419B2 (en) 2008-09-02 2013-09-24 Actimis Pharmaceuticals, Inc. Isotopically enriched pyrimidin-5-yl acetic acid derivatives as CRTH2 antagonists
WO2010027448A1 (en) * 2008-09-02 2010-03-11 Actimis Pharmaceuticals Inc. Isotopically enriched pyrimidin-5-yl acetic acid derivatives as crth2 antagonists
EP2161259A1 (en) * 2008-09-03 2010-03-10 Bayer CropScience AG 4-Haloalkyl substituted Diaminopyrimidine
WO2010025831A1 (en) * 2008-09-03 2010-03-11 Bayer Cropscience Ag Heterocyclically substituted anilinopyrimidines as fungicides
WO2010025833A1 (en) * 2008-09-03 2010-03-11 Bayer Cropscience Ag Alkoxy-substituted and alkylthio-substituted anilinopyrimidines
WO2010025871A1 (en) * 2008-09-03 2010-03-11 Bayer Cropscience Ag 4-halogen alkyl-substituted diaminopyrimidines for use as fungicides
EP2179991A1 (en) 2008-10-21 2010-04-28 Bayer Schering Pharma Aktiengesellschaft Sulfoximine substituted aniline pyrimidine derivatives as CDK inhibitors, their manufacture and use as medicine
EP2179992A1 (en) 2008-10-21 2010-04-28 Bayer Schering Pharma Aktiengesellschaft Sulfon substituted aniline pyrimidine derivatives as CDK inhibitors, their manufacture and use as medicine
US8377943B2 (en) 2008-11-14 2013-02-19 Boehringer Ingelheim International Gmbh 2,4-diaminopyrimidine derivates as PTK2-inhibitors for the treatment of abnormal cell growth
US8785464B2 (en) 2008-11-24 2014-07-22 Boehringer Ingelheim International Gmbh Pyrimidine derivatives that inhibit FAK/PTK2
US8846689B2 (en) 2008-11-24 2014-09-30 Boehringer Ingelheim International Gmbh Substituted pyrimidines for the treatment of diseases such as cancer
WO2010058030A1 (en) * 2008-11-24 2010-05-27 Boehringer Ingelheim International Gmbh New compounds
US9676762B2 (en) 2008-11-24 2017-06-13 Boehringer Ingelheim International Gmbh Pyrimidine compounds containing seven-membered fused ring systems
DE102009001438A1 (en) 2009-03-10 2010-09-16 Bayer Schering Pharma Aktiengesellschaft New carbonylamino-substituted anilino-pyrimidine compounds are tyrosine kinase-2 inhibitors, useful for treating diseases associated with inflammatory conditions e.g. bronchitis, rheumatoid arthritis, psoriasis and Guillain Barre syndrome
DE102009015070A1 (en) 2009-03-30 2010-10-14 Bayer Schering Pharma Aktiengesellschaft New phenyl-pyrimidin-2-yl-amine compounds are tyrosine kinase 2 inhibitors useful for treating e.g. rheumatoid arthritis, Crohn's disease, asthma, multiple sclerosis, adult respiratory distress syndrome, allergic alveolitis and uveitis
US8258316B2 (en) 2009-06-08 2012-09-04 Gilead Sciences, Inc. Alkanoylamino benzamide aniline HDAC inhibitor compounds
US8283357B2 (en) 2009-06-08 2012-10-09 Gilead Sciences, Inc. Cycloalkylcarbamate benzamide aniline HDAC inhibitor compounds
US8933227B2 (en) 2009-08-14 2015-01-13 Boehringer Ingelheim International Gmbh Selective synthesis of functionalized pyrimidines
US8729265B2 (en) 2009-08-14 2014-05-20 Boehringer Ingelheim International Gmbh Regioselective preparation of 2-amino-5-trifluoromethylpyrimidine derivatives
JP2013501758A (en) * 2009-08-14 2013-01-17 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Regioselective preparation of 2-amino-5-trifluoromethylpyrimidine derivatives
WO2011018518A1 (en) 2009-08-14 2011-02-17 Boehringer Ingelheim International Gmbh Regioselective preparation of 2 -amino-5-trifluoromethylpyrimidine derivatives
WO2011018517A1 (en) 2009-08-14 2011-02-17 Boehringer Ingelheim International Gmbh Regioselective preparation of 2-amino-5-trifluoromethylpyrimidine derivatives
US9556426B2 (en) 2009-09-16 2017-01-31 Celgene Avilomics Research, Inc. Protein kinase conjugates and inhibitors
US10662195B2 (en) 2009-09-16 2020-05-26 Celgene Car Llc Protein kinase conjugates and inhibitors
EP2519664A1 (en) * 2009-12-30 2012-11-07 Avila Therapeutics, Inc. Ligand-directed covalent modification of protein
EP2519664A4 (en) * 2009-12-30 2014-03-12 Avila Therapeutics Inc Ligand-directed covalent modification of protein
US11542492B2 (en) 2009-12-30 2023-01-03 Celgene Car Llc Ligand-directed covalent modification of protein
US8354420B2 (en) 2010-06-04 2013-01-15 Genentech, Inc. Aminopyrimidine derivatives as LRRK2 inhibitors
US8815882B2 (en) 2010-11-10 2014-08-26 Genentech, Inc. Pyrazole aminopyrimidine derivatives as LRRK2 modulators
US20120329771A1 (en) * 2010-11-18 2012-12-27 Boehringer Ingelheim International Gmbh Benzylic oxindole pyrimidines
US8546443B2 (en) * 2010-12-21 2013-10-01 Boehringer Ingelheim International Gmbh Benzylic oxindole pyrimidines
US10813930B2 (en) 2010-12-22 2020-10-27 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US9533954B2 (en) 2010-12-22 2017-01-03 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US10213427B2 (en) 2010-12-22 2019-02-26 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US9610289B2 (en) 2011-04-01 2017-04-04 Genentech, Inc. Combinations of AKT inhibitor compounds and erlotinib, and methods of use
US9346789B2 (en) 2011-04-01 2016-05-24 Genentech, Inc. Combinations of AKT inhibitor compounds and abiraterone, and methods of use
US9150549B2 (en) 2011-04-01 2015-10-06 Genentech, Inc. Combinations of AKT inhibitor compounds and erlotinib, and methods of use
US9682082B2 (en) 2011-04-01 2017-06-20 Genentech, Inc. Combinations of AKT and MEK inhibitor compounds, and methods of use
US9150548B2 (en) 2011-04-01 2015-10-06 Genentech, Inc. Combinations of AKT inhibitor compounds and vemurafenib, and methods of use
US9717730B2 (en) 2011-04-01 2017-08-01 Genentech, Inc. Combinations of AKT inhibitor compounds and chemotherapeutic agents, and methods of use
US10092567B2 (en) 2011-04-01 2018-10-09 Genentech, Inc. Combinations of AKT inhibitor compounds and chemotherapeutic agents, and methods of use
WO2013041537A1 (en) 2011-09-23 2013-03-28 Beiersdorf Ag Heterocyclocarbonylaminothiazoles, cosmetic or dermatological preparations containing said heterocyclocarbonylaminothiazoles, and use thereof to combat or prevent undesired pigmentation of the skin
DE102011083283A1 (en) 2011-09-23 2013-03-28 Beiersdorf Ag Heteroalkylamidothiazoles, their cosmetic or dermatological use and cosmetic or dermatological preparations containing such Heteroalkylamidothiazolen
US10233157B2 (en) 2012-03-13 2019-03-19 Dana-Farber Cancer Institute, Inc. SIK inhibitor for use in a method of treating an inflammatory and/or immune disorder
WO2013157540A1 (en) * 2012-04-17 2013-10-24 富士フイルム株式会社 Nitrogen-containing heterocyclic compound or salt thereof
AU2013250378B2 (en) * 2012-04-17 2016-01-14 Fujifilm Corporation Nitrogen-containing heterocyclic compound or salt thereof
US9611267B2 (en) 2012-06-13 2017-04-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US11053246B2 (en) 2012-06-13 2021-07-06 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US10131667B2 (en) 2012-06-13 2018-11-20 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US11840534B2 (en) 2012-06-13 2023-12-12 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US9388185B2 (en) 2012-08-10 2016-07-12 Incyte Holdings Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9745311B2 (en) 2012-08-10 2017-08-29 Incyte Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
US9533984B2 (en) 2013-04-19 2017-01-03 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10450313B2 (en) 2013-04-19 2019-10-22 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US11530214B2 (en) 2013-04-19 2022-12-20 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10947230B2 (en) 2013-04-19 2021-03-16 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US10040790B2 (en) 2013-04-19 2018-08-07 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
EP3030241A4 (en) * 2013-08-06 2019-06-19 H. Lee Moffitt Cancer Center And Research Institute, Inc. Inhibitors of ack1/tnk2 tyrosine kinase
US10975058B2 (en) 2014-07-21 2021-04-13 Dana-Farber Cancer Institute, Inc. Imidazolyl kinase inhibitors and uses thereof
US10287268B2 (en) 2014-07-21 2019-05-14 Dana-Farber Cancer Institute, Inc. Imidazolyl kinase inhibitors and uses thereof
US10457691B2 (en) 2014-07-21 2019-10-29 Dana-Farber Cancer Institute, Inc. Macrocyclic kinase inhibitors and uses thereof
US10265321B2 (en) 2014-08-08 2019-04-23 Dana-Farber Cancer Institute, Inc. Uses of salt-inducible kinase (SIK) inhibitors
EP3177295A4 (en) * 2014-08-08 2018-06-13 Dana Farber Cancer Institute, Inc. Uses of salt-inducible kinase (sik) inhibitors
US10011571B2 (en) 2014-09-16 2018-07-03 Shenzhen Chipscreen Biosciences, Ltd. Preparation method for aromatic heterocyclic compound used as selective JAK3 and/or JAK1 kinase inhibitor and application of aromatic heterocyclic compound
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10016438B2 (en) 2015-02-20 2018-07-10 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9801889B2 (en) 2015-02-20 2017-10-31 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9580423B2 (en) 2015-02-20 2017-02-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10214528B2 (en) 2015-02-20 2019-02-26 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10632126B2 (en) 2015-02-20 2020-04-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11014923B2 (en) 2015-02-20 2021-05-25 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10251892B2 (en) 2015-02-20 2019-04-09 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11173162B2 (en) 2015-02-20 2021-11-16 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10738048B2 (en) 2015-02-20 2020-08-11 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11667635B2 (en) 2015-02-20 2023-06-06 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10954242B2 (en) 2016-07-05 2021-03-23 The Broad Institute, Inc. Bicyclic urea kinase inhibitors and uses thereof
US11725011B2 (en) 2016-07-05 2023-08-15 The General Hospital Corporation Bicyclic urea kinase inhibitors and uses thereof
US11241435B2 (en) 2016-09-16 2022-02-08 The General Hospital Corporation Uses of salt-inducible kinase (SIK) inhibitors for treating osteoporosis
US11285158B2 (en) 2017-02-28 2022-03-29 The General Hospital Corporation Uses of pyrimidopyrimidinones as SIK inhibitors
US11878019B2 (en) 2017-02-28 2024-01-23 The General Hospital Corporation Uses of pyrimidopyrimidinones as SIK inhibitors
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US11472801B2 (en) 2017-05-26 2022-10-18 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
WO2020023851A1 (en) * 2018-07-26 2020-01-30 Yale University Bifunctional substitued pyrimidines as modulators of fak proteolyse
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
CN114127057A (en) * 2019-05-10 2022-03-01 德西费拉制药有限责任公司 Phenylaminopyrimidine amide autophagy inhibitors and methods of use thereof
WO2020231806A1 (en) * 2019-05-10 2020-11-19 Deciphera Pharmaceuticals, Llc Phenylaminopyrimidine amide autophagy inhibitors and methods of use thereof
US11591329B2 (en) 2019-07-09 2023-02-28 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors

Also Published As

Publication number Publication date
HRP20050601A2 (en) 2005-10-31
MXPA05005547A (en) 2005-07-26
JP2006508997A (en) 2006-03-16
PL377795A1 (en) 2006-02-20
EA200500721A1 (en) 2005-12-29
IL168102A0 (en) 2009-02-11
NZ539823A (en) 2008-04-30
BR0316680A (en) 2005-10-18
IS7881A (en) 2005-06-02
US20040186118A1 (en) 2004-09-23
US7504410B2 (en) 2009-03-17
KR20050084027A (en) 2005-08-26
NO20053144D0 (en) 2005-06-27
CN1717396A (en) 2006-01-04
EP1565446A1 (en) 2005-08-24
RS20050363A (en) 2007-11-15
AU2003288198A1 (en) 2004-06-18
CA2502970A1 (en) 2004-06-10
NO20053144L (en) 2005-06-27
ECSP055899A (en) 2005-11-22
ZA200505184B (en) 2006-09-27

Similar Documents

Publication Publication Date Title
US7504410B2 (en) Chk-, Pdk- and Akt-inhibitory pyrimidines, their production and use as pharmaceutical agents
US7235561B2 (en) Compound and a composition including such a compound
JP4700006B2 (en) Sulfoximine-substituted pyrimidines as CDK- and / or VEGF inhibitors, their production and use as pharmaceuticals
US8133900B2 (en) Use of bi-aryl meta-pyrimidine inhibitors of kinases
RU2597364C2 (en) Bi-aryl-meta-pyrimidine kinase inhibitors
CZ2003617A3 (en) Imidazol-5-yl-2-anilinopyrimidine derivatives, process of their preparation and pharmaceutical preparation in which they are comprised
US20040209895A1 (en) Macrocyclic pyrimidines, their production and use as pharmaceutical agents
US7288547B2 (en) CDK-inhibitory 2-heteroaryl-pyrimidines, their production and use as pharmaceutical agents
US20110212077A1 (en) Bi-aryl meta-pyrimidine inhibitors of kinases
US20050090541A1 (en) Indolinone derivatives and their use in treating disease-states such as cancer
EP1705177A1 (en) N-aryl-sulfoximine-substituted pyrimidines as CDK- and/or VEGF inhibitors, their production and use as pharmaceutical agents
KR20040041092A (en) Use of gal3 receptor antagonists for the treatment of depression and/or anxiety and compounds useful in such methods

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: P-2005/0363

Country of ref document: YU

AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 168102

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2502970

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1603/DELNP/2005

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2003780086

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 539823

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 377795

Country of ref document: PL

WWE Wipo information: entry into national phase

Ref document number: PA/a/2005/005547

Country of ref document: MX

Ref document number: 200500721

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 1-2005-501019

Country of ref document: PH

Ref document number: 2004554522

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 1020057009682

Country of ref document: KR

Ref document number: 2003288198

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 20038A45442

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: CR2005-007866

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: 05061944

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 2005/05184

Country of ref document: ZA

Ref document number: 200505184

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: P20050601A

Country of ref document: HR

WWP Wipo information: published in national office

Ref document number: 2003780086

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020057009682

Country of ref document: KR

ENP Entry into the national phase

Ref document number: PI0316680

Country of ref document: BR